An anterior pituitary hormone that stimulates the ADRENAL CORTEX and its production of CORTICOSTEROIDS. ACTH is a 39-amino acid polypeptide of which the N-terminal 24-amino acid segment is identical in all species and contains the adrenocorticotrophic activity. Upon further tissue-specific processing, ACTH can yield ALPHA-MSH and corticotrophin-like intermediate lobe peptide (CLIP).
The main glucocorticoid secreted by the ADRENAL CORTEX. Its synthetic counterpart is used, either as an injection or topically, in the treatment of inflammation, allergy, collagen diseases, asthma, adrenocortical deficiency, shock, and some neoplastic conditions.
A pair of glands located at the cranial pole of each of the two KIDNEYS. Each adrenal gland is composed of two distinct endocrine tissues with separate embryonic origins, the ADRENAL CORTEX producing STEROIDS and the ADRENAL MEDULLA producing NEUROTRANSMITTERS.
The interactions between the anterior pituitary and adrenal glands, in which corticotropin (ACTH) stimulates the adrenal cortex and adrenal cortical hormones suppress the production of corticotropin by the anterior pituitary.
Chemical substances having a specific regulatory effect on the activity of a certain organ or organs. The term was originally applied to substances secreted by various ENDOCRINE GLANDS and transported in the bloodstream to the target organs. It is sometimes extended to include those substances that are not produced by the endocrine glands but that have similar effects.
A collection of NEURONS, tracts of NERVE FIBERS, endocrine tissue, and blood vessels in the HYPOTHALAMUS and the PITUITARY GLAND. This hypothalamo-hypophyseal portal circulation provides the mechanism for hypothalamic neuroendocrine (HYPOTHALAMIC HORMONES) regulation of pituitary function and the release of various PITUITARY HORMONES into the systemic circulation to maintain HOMEOSTASIS.
A peptide of about 41 amino acids that stimulates the release of ADRENOCORTICOTROPIC HORMONE. CRH is synthesized by neurons in the PARAVENTRICULAR NUCLEUS of the HYPOTHALAMUS. After being released into the pituitary portal circulation, CRH stimulates the release of ACTH from the PITUITARY GLAND. CRH can also be synthesized in other tissues, such as PLACENTA; ADRENAL MEDULLA; and TESTIS.
An epileptic syndrome characterized by the triad of infantile spasms, hypsarrhythmia, and arrest of psychomotor development at seizure onset. The majority present between 3-12 months of age, with spasms consisting of combinations of brief flexor or extensor movements of the head, trunk, and limbs. The condition is divided into two forms: cryptogenic (idiopathic) and symptomatic (secondary to a known disease process such as intrauterine infections; nervous system abnormalities; BRAIN DISEASES, METABOLIC, INBORN; prematurity; perinatal asphyxia; TUBEROUS SCLEROSIS; etc.). (From Menkes, Textbook of Child Neurology, 5th ed, pp744-8)
An adrenocortical steroid that has modest but significant activities as a mineralocorticoid and a glucocorticoid. (From Goodman and Gilman's The Pharmacological Basis of Therapeutics, 8th ed, p1437)
The outer layer of the adrenal gland. It is derived from MESODERM and comprised of three zones (outer ZONA GLOMERULOSA, middle ZONA FASCICULATA, and inner ZONA RETICULARIS) with each producing various steroids preferentially, such as ALDOSTERONE; HYDROCORTISONE; DEHYDROEPIANDROSTERONE; and ANDROSTENEDIONE. Adrenal cortex function is regulated by pituitary ADRENOCORTICOTROPIN.
A condition caused by prolonged exposure to excess levels of cortisol (HYDROCORTISONE) or other GLUCOCORTICOIDS from endogenous or exogenous sources. It is characterized by upper body OBESITY; OSTEOPOROSIS; HYPERTENSION; DIABETES MELLITUS; HIRSUTISM; AMENORRHEA; and excess body fluid. Endogenous Cushing syndrome or spontaneous hypercortisolism is divided into two groups, those due to an excess of ADRENOCORTICOTROPIN and those that are ACTH-independent.
Conditions in which the production of adrenal CORTICOSTEROIDS falls below the requirement of the body. Adrenal insufficiency can be caused by defects in the ADRENAL GLANDS, the PITUITARY GLAND, or the HYPOTHALAMUS.
Cell surface receptors that bind CORTICOTROPIN; (ACTH, adrenocorticotropic hormone) with high affinity and trigger intracellular changes. Pharmacology suggests there may be multiple ACTH receptors. An ACTH receptor has been cloned and belongs to a subfamily of G-protein-coupled receptors. In addition to the adrenal cortex, ACTH receptors are found in the brain and immune systems.
A small, unpaired gland situated in the SELLA TURCICA. It is connected to the HYPOTHALAMUS by a short stalk which is called the INFUNDIBULUM.
Peptides with the ability to stimulate pigmented cells MELANOCYTES in mammals and MELANOPHORES in lower vertebrates. By stimulating the synthesis and distribution of MELANIN in these pigmented cells, they increase coloration of skin and other tissue. MSHs, derived from pro-opiomelanocortin (POMC), are produced by MELANOTROPHS in the INTERMEDIATE LOBE OF PITUITARY; CORTICOTROPHS in the ANTERIOR LOBE OF PITUITARY, and the hypothalamic neurons in the ARCUATE NUCLEUS OF HYPOTHALAMUS.
A synthetic peptide that is identical to the 24-amino acid segment at the N-terminal of ADRENOCORTICOTROPIC HORMONE. ACTH (1-24), a segment similar in all species, contains the biological activity that stimulates production of CORTICOSTEROIDS in the ADRENAL CORTEX.
A melanocortin receptor subtype found primarily in the ADRENAL CORTEX. It shows specificity for ADRENOCORTICOTROPIC HORMONE.
Excess production of ADRENAL CORTEX HORMONES such as ALDOSTERONE; HYDROCORTISONE; DEHYDROEPIANDROSTERONE; and/or ANDROSTENEDIONE. Hyperadrenal syndromes include CUSHING SYNDROME; HYPERALDOSTERONISM; and VIRILISM.
A 30-kDa protein synthesized primarily in the ANTERIOR PITUITARY GLAND and the HYPOTHALAMUS. It is also found in the skin and other peripheral tissues. Depending on species and tissues, POMC is cleaved by PROHORMONE CONVERTASES yielding various active peptides including ACTH; BETA-LIPOTROPIN; ENDORPHINS; MELANOCYTE-STIMULATING HORMONES; and others (GAMMA-LPH; CORTICOTROPIN-LIKE INTERMEDIATE LOBE PEPTIDE; N-terminal peptide of POMC or NPP).
A major gonadotropin secreted by the adenohypophysis (PITUITARY GLAND, ANTERIOR). Follicle-stimulating hormone stimulates GAMETOGENESIS and the supporting cells such as the ovarian GRANULOSA CELLS, the testicular SERTOLI CELLS, and LEYDIG CELLS. FSH consists of two noncovalently linked subunits, alpha and beta. Within a species, the alpha subunit is common in the three pituitary glycoprotein hormones (TSH, LH, and FSH), but the beta subunit is unique and confers its biological specificity.
A major gonadotropin secreted by the adenohypophysis (PITUITARY GLAND, ANTERIOR). Luteinizing hormone regulates steroid production by the interstitial cells of the TESTIS and the OVARY. The preovulatory LUTEINIZING HORMONE surge in females induces OVULATION, and subsequent LUTEINIZATION of the follicle. LUTEINIZING HORMONE consists of two noncovalently linked subunits, alpha and beta. Within a species, the alpha subunit is common in the three pituitary glycoprotein hormones (TSH, LH and FSH), but the beta subunit is unique and confers its biological specificity.
Hormones secreted by the PITUITARY GLAND including those from the anterior lobe (adenohypophysis), the posterior lobe (neurohypophysis), and the ill-defined intermediate lobe. Structurally, they include small peptides, proteins, and glycoproteins. They are under the regulation of neural signals (NEUROTRANSMITTERS) or neuroendocrine signals (HYPOTHALAMIC HORMONES) from the hypothalamus as well as feedback from their targets such as ADRENAL CORTEX HORMONES; ANDROGENS; ESTROGENS.
Symptom complex due to ACTH production by non-pituitary neoplasms.
Neoplasms which arise from or metastasize to the PITUITARY GLAND. The majority of pituitary neoplasms are adenomas, which are divided into non-secreting and secreting forms. Hormone producing forms are further classified by the type of hormone they secrete. Pituitary adenomas may also be characterized by their staining properties (see ADENOMA, BASOPHIL; ADENOMA, ACIDOPHIL; and ADENOMA, CHROMOPHOBE). Pituitary tumors may compress adjacent structures, including the HYPOTHALAMUS, several CRANIAL NERVES, and the OPTIC CHIASM. Chiasmal compression may result in bitemporal HEMIANOPSIA.
Natural hormones secreted by the THYROID GLAND, such as THYROXINE, and their synthetic analogs.
A 31-amino acid peptide that is the C-terminal fragment of BETA-LIPOTROPIN. It acts on OPIOID RECEPTORS and is an analgesic. Its first four amino acids at the N-terminal are identical to the tetrapeptide sequence of METHIONINE ENKEPHALIN and LEUCINE ENKEPHALIN.
Use of a device for the purpose of controlling movement of all or part of the body. Splinting and casting are FRACTURE FIXATION.
Hormones secreted by the adenohypophysis (PITUITARY GLAND, ANTERIOR). Structurally, they include polypeptide, protein, and glycoprotein molecules.
An inhibitor of the enzyme STEROID 11-BETA-MONOOXYGENASE. It is used as a test of the feedback hypothalamic-pituitary mechanism in the diagnosis of CUSHING SYNDROME.
Surgical removal or destruction of the hypophysis, or pituitary gland. (Dorland, 28th ed)
The anterior glandular lobe of the pituitary gland, also known as the adenohypophysis. It secretes the ADENOHYPOPHYSEAL HORMONES that regulate vital functions such as GROWTH; METABOLISM; and REPRODUCTION.
Examinations that evaluate and monitor hormone production in the adrenal cortex.
A general state of sluggishness, listless, or uninterested, with being tired, and having difficulty concentrating and doing simple tasks. It may be related to DEPRESSION or DRUG ADDICTION.
A disease of the PITUITARY GLAND characterized by the excess amount of ADRENOCORTICOTROPIC HORMONE secreted. This leads to hypersecretion of cortisol (HYDROCORTISONE) by the ADRENAL GLANDS resulting in CUSHING SYNDROME.
A 13-amino acid peptide derived from proteolytic cleavage of ADRENOCORTICOTROPIC HORMONE, the N-terminal segment of ACTH. ACTH (1-13) is amidated at the C-terminal to form ACTH (1-13)NH2 which in turn is acetylated to form alpha-MSH in the secretory granules. Alpha-MSH stimulates the synthesis and distribution of MELANIN in MELANOCYTES in mammals and MELANOPHORES in lower vertebrates.
The unfavorable effect of environmental factors (stressors) on the physiological functions of an organism. Prolonged unresolved physiological stress can affect HOMEOSTASIS of the organism, and may lead to damaging or pathological conditions.
Excision of one or both adrenal glands. (From Dorland, 28th ed)
A decapeptide that stimulates the synthesis and secretion of both pituitary gonadotropins, LUTEINIZING HORMONE and FOLLICLE STIMULATING HORMONE. GnRH is produced by neurons in the septum PREOPTIC AREA of the HYPOTHALAMUS and released into the pituitary portal blood, leading to stimulation of GONADOTROPHS in the ANTERIOR PITUITARY GLAND.
The wide middle zone of the adrenal cortex. This zone produces a series of enzymes that convert PREGNENOLONE to cortisol (HYDROCORTISONE) via 17-ALPHA-HYDROXYPROGESTERONE.
A group of CORTICOSTEROIDS that affect carbohydrate metabolism (GLUCONEOGENESIS, liver glycogen deposition, elevation of BLOOD SUGAR), inhibit ADRENOCORTICOTROPIC HORMONE secretion, and possess pronounced anti-inflammatory activity. They also play a role in fat and protein metabolism, maintenance of arterial blood pressure, alteration of the connective tissue response to injury, reduction in the number of circulating lymphocytes, and functioning of the central nervous system.
A general term collectively applied to tumors associated with the APUD CELLS series, irrespective of their specific identification.
Tests that evaluate the adrenal glands controlled by pituitary hormones.
A group of corticosteroids bearing a hydroxy group at the 11-position.
A polypeptide hormone (84 amino acid residues) secreted by the PARATHYROID GLANDS which performs the essential role of maintaining intracellular CALCIUM levels in the body. Parathyroid hormone increases intracellular calcium by promoting the release of CALCIUM from BONE, increases the intestinal absorption of calcium, increases the renal tubular reabsorption of calcium, and increases the renal excretion of phosphates.
A family of G-protein-coupled receptors that have specificity for MELANOCYTE-STIMULATING HORMONES and ADRENOCORTICOTROPIC HORMONE. There are several subtypes of melanocortin receptors, each having a distinct ligand specificity profile and tissue localization.
A 191-amino acid polypeptide hormone secreted by the human adenohypophysis (PITUITARY GLAND, ANTERIOR), also known as GH or somatotropin. Synthetic growth hormone, termed somatropin, has replaced the natural form in therapeutic usage such as treatment of dwarfism in children with growth hormone deficiency.
A lactogenic hormone secreted by the adenohypophysis (PITUITARY GLAND, ANTERIOR). It is a polypeptide of approximately 23 kD. Besides its major action on lactation, in some species prolactin exerts effects on reproduction, maternal behavior, fat metabolism, immunomodulation and osmoregulation. Prolactin receptors are present in the mammary gland, hypothalamus, liver, ovary, testis, and prostate.
A metabolite of PROGESTERONE with a hydroxyl group at the 17-alpha position. It serves as an intermediate in the biosynthesis of HYDROCORTISONE and GONADAL STEROID HORMONES.
Diminution or cessation of secretion of one or more hormones from the anterior pituitary gland (including LH; FOLLICLE STIMULATING HORMONE; SOMATOTROPIN; and CORTICOTROPIN). This may result from surgical or radiation ablation, non-secretory PITUITARY NEOPLASMS, metastatic tumors, infarction, PITUITARY APOPLEXY, infiltrative or granulomatous processes, and other conditions.
Examinations that evaluate functions of the pituitary gland.
Steroid hormones produced by the GONADS. They stimulate reproductive organs, germ cell maturation, and the secondary sex characteristics in the males and the females. The major sex steroid hormones include ESTRADIOL; PROGESTERONE; and TESTOSTERONE.
Classic quantitative assay for detection of antigen-antibody reactions using a radioactively labeled substance (radioligand) either directly or indirectly to measure the binding of the unlabeled substance to a specific antibody or other receptor system. Non-immunogenic substances (e.g., haptens) can be measured if coupled to larger carrier proteins (e.g., bovine gamma-globulin or human serum albumin) capable of inducing antibody formation.
A hormone secreted by the ADRENAL CORTEX that regulates electrolyte and water balance by increasing the renal retention of sodium and the excretion of potassium.
An anti-inflammatory 9-fluoro-glucocorticoid.
A naturally occurring glucocorticoid. It has been used in replacement therapy for adrenal insufficiency and as an anti-inflammatory agent. Cortisone itself is inactive. It is converted in the liver to the active metabolite HYDROCORTISONE. (From Martindale, The Extra Pharmacopoeia, 30th ed, p726)
The intermediate lobe of the pituitary gland. It shows considerable size variation among the species, small in humans, and large in amphibians and lower vertebrates. This lobe produces mainly MELANOCYTE-STIMULATING HORMONES and other peptides from post-translational processing of pro-opiomelanocortin (POMC).
Anterior pituitary cells that produce ADRENOCORTICOTROPHIC HORMONE.
Adrenal cortex hormones are steroid hormones produced by the outer portion of the adrenal gland, consisting of glucocorticoids, mineralocorticoids, and androgens, which play crucial roles in various physiological processes such as metabolism regulation, stress response, electrolyte balance, and sexual development and function.
A glycoprotein hormone secreted by the adenohypophysis (PITUITARY GLAND, ANTERIOR). Thyrotropin stimulates THYROID GLAND by increasing the iodide transport, synthesis and release of thyroid hormones (THYROXINE and TRIIODOTHYRONINE). Thyrotropin consists of two noncovalently linked subunits, alpha and beta. Within a species, the alpha subunit is common in the pituitary glycoprotein hormones (TSH; LUTEINIZING HORMONE and FSH), but the beta subunit is unique and confers its biological specificity.
Sampling of blood levels of the adrenocorticotropic hormone (ACTH) by withdrawal of blood from the inferior petrosal sinus. The inferior petrosal sinus arises from the cavernous sinus and runs to the internal jugular vein. Sampling of blood at this level is a valuable tool in the differential diagnosis of Cushing disease, Cushing syndrome, and other adrenocortical diseases.
Ventral part of the DIENCEPHALON extending from the region of the OPTIC CHIASM to the caudal border of the MAMMILLARY BODIES and forming the inferior and lateral walls of the THIRD VENTRICLE.
Nucleus in the anterior part of the HYPOTHALAMUS.
Specific high affinity binding proteins for THYROID HORMONES in target cells. They are usually found in the nucleus and regulate DNA transcription. These receptors are activated by hormones that leads to transcription, cell differentiation, and growth suppression. Thyroid hormone receptors are encoded by two genes (GENES, ERBA): erbA-alpha and erbA-beta for alpha and beta thyroid hormone receptors, respectively.
A pituitary adenoma which secretes ADRENOCORTICOTROPIN, leading to CUSHING DISEASE.
Pathological processes of the ADRENAL GLANDS.
A system of NEURONS that has the specialized function to produce and secrete HORMONES, and that constitutes, in whole or in part, an ENDOCRINE SYSTEM or organ.
A major C19 steroid produced by the ADRENAL CORTEX. It is also produced in small quantities in the TESTIS and the OVARY. Dehydroepiandrosterone (DHEA) can be converted to TESTOSTERONE; ANDROSTENEDIONE; ESTRADIOL; and ESTRONE. Most of DHEA is sulfated (DEHYDROEPIANDROSTERONE SULFATE) before secretion.
The predominant form of mammalian antidiuretic hormone. It is a nonapeptide containing an ARGININE at residue 8 and two disulfide-linked cysteines at residues of 1 and 6. Arg-vasopressin is used to treat DIABETES INSIPIDUS or to improve vasomotor tone and BLOOD PRESSURE.
A specific blocker of dopamine receptors. It speeds gastrointestinal peristalsis, causes prolactin release, and is used as antiemetic and tool in the study of dopaminergic mechanisms.
A benign epithelial tumor with a glandular organization.
Tumors or cancers of the ADRENAL CORTEX.
Stress wherein emotional factors predominate.
A group of polycyclic compounds closely related biochemically to TERPENES. They include cholesterol, numerous hormones, precursors of certain vitamins, bile acids, alcohols (STEROLS), and certain natural drugs and poisons. Steroids have a common nucleus, a fused, reduced 17-carbon atom ring system, cyclopentanoperhydrophenanthrene. Most steroids also have two methyl groups and an aliphatic side-chain attached to the nucleus. (From Hawley's Condensed Chemical Dictionary, 11th ed)
An adrenal disease characterized by the progressive destruction of the ADRENAL CORTEX, resulting in insufficient production of ALDOSTERONE and HYDROCORTISONE. Clinical symptoms include ANOREXIA; NAUSEA; WEIGHT LOSS; MUSCLE WEAKNESS; and HYPERPIGMENTATION of the SKIN due to increase in circulating levels of ACTH precursor hormone which stimulates MELANOCYTES.
Tumors or cancer of the ADRENAL GLANDS.
A potent androgenic steroid and major product secreted by the LEYDIG CELLS of the TESTIS. Its production is stimulated by LUTEINIZING HORMONE from the PITUITARY GLAND. In turn, testosterone exerts feedback control of the pituitary LH and FSH secretion. Depending on the tissues, testosterone can be further converted to DIHYDROTESTOSTERONE or ESTRADIOL.
A polypeptide that is secreted by the adenohypophysis (PITUITARY GLAND, ANTERIOR). Growth hormone, also known as somatotropin, stimulates mitosis, cell differentiation and cell growth. Species-specific growth hormones have been synthesized.
A group of inherited disorders of the ADRENAL GLANDS, caused by enzyme defects in the synthesis of cortisol (HYDROCORTISONE) and/or ALDOSTERONE leading to accumulation of precursors for ANDROGENS. Depending on the hormone imbalance, congenital adrenal hyperplasia can be classified as salt-wasting, hypertensive, virilizing, or feminizing. Defects in STEROID 21-HYDROXYLASE; STEROID 11-BETA-HYDROXYLASE; STEROID 17-ALPHA-HYDROXYLASE; 3-beta-hydroxysteroid dehydrogenase (3-HYDROXYSTEROID DEHYDROGENASES); TESTOSTERONE 5-ALPHA-REDUCTASE; or steroidogenic acute regulatory protein; among others, underlie these disorders.
The major progestational steroid that is secreted primarily by the CORPUS LUTEUM and the PLACENTA. Progesterone acts on the UTERUS, the MAMMARY GLANDS and the BRAIN. It is required in EMBRYO IMPLANTATION; PREGNANCY maintenance, and the development of mammary tissue for MILK production. Progesterone, converted from PREGNENOLONE, also serves as an intermediate in the biosynthesis of GONADAL STEROID HORMONES and adrenal CORTICOSTEROIDS.
A benign tumor of the anterior pituitary in which the cells do not stain with acidic or basic dyes.
A condition caused by the overproduction of ALDOSTERONE. It is characterized by sodium retention and potassium excretion with resultant HYPERTENSION and HYPOKALEMIA.
The major hormone derived from the thyroid gland. Thyroxine is synthesized via the iodination of tyrosines (MONOIODOTYROSINE) and the coupling of iodotyrosines (DIIODOTYROSINE) in the THYROGLOBULIN. Thyroxine is released from thyroglobulin by proteolysis and secreted into the blood. Thyroxine is peripherally deiodinated to form TRIIODOTHYRONINE which exerts a broad spectrum of stimulatory effects on cell metabolism.
The circulating form of a major C19 steroid produced primarily by the ADRENAL CORTEX. DHEA sulfate serves as a precursor for TESTOSTERONE; ANDROSTENEDIONE; ESTRADIOL; and ESTRONE.
17,21-Dihydroxypregn-4-ene-3,20-dione. A 17-hydroxycorticosteroid with glucocorticoid and anti-inflammatory activities.
Elements of limited time intervals, contributing to particular results or situations.
Antidiuretic hormones released by the NEUROHYPOPHYSIS of all vertebrates (structure varies with species) to regulate water balance and OSMOLARITY. In general, vasopressin is a nonapeptide consisting of a six-amino-acid ring with a cysteine 1 to cysteine 6 disulfide bridge or an octapeptide containing a CYSTINE. All mammals have arginine vasopressin except the pig with a lysine at position 8. Vasopressin, a vasoconstrictor, acts on the KIDNEY COLLECTING DUCTS to increase water reabsorption, increase blood volume and blood pressure.
The 17-beta-isomer of estradiol, an aromatized C18 steroid with hydroxyl group at 3-beta- and 17-beta-position. Estradiol-17-beta is the most potent form of mammalian estrogenic steroids.
The relationship between the dose of an administered drug and the response of the organism to the drug.
An involuntary contraction of a muscle or group of muscles. Spasms may involve SKELETAL MUSCLE or SMOOTH MUSCLE.
One of the three major groups of endogenous opioid peptides. They are large peptides derived from the PRO-OPIOMELANOCORTIN precursor. The known members of this group are alpha-, beta-, and gamma-endorphin. The term endorphin is also sometimes used to refer to all opioid peptides, but the narrower sense is used here; OPIOID PEPTIDES is used for the broader group.
Cell surface proteins that bind corticotropin-releasing hormone with high affinity and trigger intracellular changes which influence the behavior of cells. The corticotropin releasing-hormone receptors on anterior pituitary cells mediate the stimulation of corticotropin release by hypothalamic corticotropin releasing factor. The physiological consequence of activating corticotropin-releasing hormone receptors on central neurons is not well understood.
Peptides derived from pro-opiomelanocortin (POMC) which can stimulate MELANOCYTES or CORTICOTROPHS. Melanocortins include ACTH; ALPHA-MSH; and other peptides such as BETA-MSH and GAMMA-MSH, derived from other fragments of POMC. These peptides act through a variety of MELANOCORTIN RECEPTORS to control different functions including steroidogenesis, energy homeostasis, feeding, and skin pigmentation.
The active sympathomimetic hormone from the ADRENAL MEDULLA. It stimulates both the alpha- and beta- adrenergic systems, causes systemic VASOCONSTRICTION and gastrointestinal relaxation, stimulates the HEART, and dilates BRONCHI and cerebral vessels. It is used in ASTHMA and CARDIAC FAILURE and to delay absorption of local ANESTHETICS.
An adenine nucleotide containing one phosphate group which is esterified to both the 3'- and 5'-positions of the sugar moiety. It is a second messenger and a key intracellular regulator, functioning as a mediator of activity for a number of hormones, including epinephrine, glucagon, and ACTH.
A 90-amino acid peptide derived from post-translational processing of pro-opiomelanocortin (POMC) in the PITUITARY GLAND and the HYPOTHALAMUS. It is the C-terminal fragment of POMC with lipid-mobilizing activities, such as LIPOLYSIS and steroidogenesis. Depending on the species and the tissue sites, beta-LPH may be further processed to yield active peptides including GAMMA-LIPOTROPIN; BETA-MSH; and ENDORPHINS.
Any of the ruminant mammals with curved horns in the genus Ovis, family Bovidae. They possess lachrymal grooves and interdigital glands, which are absent in GOATS.
A group of hydroxycorticosteroids bearing a hydroxy group at the 17-position. Urinary excretion of these compounds is used as an index of adrenal function. They are used systemically in the free alcohol form, but with esterification of the hydroxy groups, topical effectiveness is increased.
Syndromes resulting from inappropriate production of HORMONES or hormone-like materials by NEOPLASMS in non-endocrine tissues or not by the usual ENDOCRINE GLANDS. Such hormone outputs are called ectopic hormone (HORMONES, ECTOPIC) secretion.
Disorders involving either the ADENOHYPOPHYSIS or the NEUROHYPOPHYSIS. These diseases usually manifest as hypersecretion or hyposecretion of PITUITARY HORMONES. Neoplastic pituitary masses can also cause compression of the OPTIC CHIASM and other adjacent structures.
A usually small, slow-growing neoplasm composed of islands of rounded, oxyphilic, or spindle-shaped cells of medium size, with moderately small vesicular nuclei, and covered by intact mucosa with a yellow cut surface. The tumor can occur anywhere in the gastrointestinal tract (and in the lungs and other sites); approximately 90% arise in the appendix. It is now established that these tumors are of neuroendocrine origin and derive from a primitive stem cell. (From Stedman, 25th ed & Holland et al., Cancer Medicine, 3d ed, p1182)
A strain of albino rat used widely for experimental purposes because of its calmness and ease of handling. It was developed by the Sprague-Dawley Animal Company.
Domesticated bovine animals of the genus Bos, usually kept on a farm or ranch and used for the production of meat or dairy products or for heavy labor.
RNA sequences that serve as templates for protein synthesis. Bacterial mRNAs are generally primary transcripts in that they do not require post-transcriptional processing. Eukaryotic mRNA is synthesized in the nucleus and must be exported to the cytoplasm for translation. Most eukaryotic mRNAs have a sequence of polyadenylic acid at the 3' end, referred to as the poly(A) tail. The function of this tail is not known for certain, but it may play a role in the export of mature mRNA from the nucleus as well as in helping stabilize some mRNA molecules by retarding their degradation in the cytoplasm.
A malignant olfactory neuroblastoma arising from the olfactory epithelium of the superior nasal cavity and cribriform plate. It is uncommon (3% of nasal tumors) and rarely is associated with the production of excess hormones (e.g., SIADH, Cushing Syndrome). It has a high propensity for multiple local recurrences and bony metastases. (From Holland et al., Cancer Medicine, 3rd ed, p1245; J Laryngol Otol 1998 Jul;112(7):628-33)
A mitochondrial cytochrome P450 enzyme that catalyzes the 11-beta-hydroxylation of steroids in the presence of molecular oxygen and NADPH-FERRIHEMOPROTEIN REDUCTASE. This enzyme, encoded by CYP11B1 gene, is important in the synthesis of CORTICOSTERONE and HYDROCORTISONE. Defects in CYP11B1 cause congenital adrenal hyperplasia (ADRENAL HYPERPLASIA, CONGENITAL).
Cytoplasmic proteins that specifically bind glucocorticoids and mediate their cellular effects. The glucocorticoid receptor-glucocorticoid complex acts in the nucleus to induce transcription of DNA. Glucocorticoids were named for their actions on blood glucose concentration, but they have equally important effects on protein and fat metabolism. Cortisol is the most important example.
A T3 thyroid hormone normally synthesized and secreted by the thyroid gland in much smaller quantities than thyroxine (T4). Most T3 is derived from peripheral monodeiodination of T4 at the 5' position of the outer ring of the iodothyronine nucleus. The hormone finally delivered and used by the tissues is mainly T3.
A pituitary adenoma which secretes PROLACTIN, leading to HYPERPROLACTINEMIA. Clinical manifestations include AMENORRHEA; GALACTORRHEA; IMPOTENCE; HEADACHE; visual disturbances; and CEREBROSPINAL FLUID RHINORRHEA.
Hormones released from neoplasms or from other cells that are not the usual sources of hormones.
Compounds, either natural or synthetic, which block development of the growing insect.
A statistical technique that isolates and assesses the contributions of categorical independent variables to variation in the mean of a continuous dependent variable.
The status during which female mammals carry their developing young (EMBRYOS or FETUSES) in utero before birth, beginning from FERTILIZATION to BIRTH.
The regular recurrence, in cycles of about 24 hours, of biological processes or activities, such as sensitivity to drugs and stimuli, hormone secretion, sleeping, and feeding.
A serpin family member that binds to and transports GLUCOCORTICOIDS in the BLOOD.
Arthritis, especially of the great toe, as a result of gout. Acute gouty arthritis often is precipitated by trauma, infection, surgery, etc. The initial attacks are usually monoarticular but later attacks are often polyarticular.
A plant genus of the LAMIACEAE family.
Therapeutic use of hormones to alleviate the effects of hormone deficiency.
The narrow subcapsular outer zone of the adrenal cortex. This zone produces a series of enzymes that convert PREGNENOLONE to ALDOSTERONE. The final steps involve three successive oxidations by CYTOCHROME P-450 CYP11B2.
Ductless glands that secrete HORMONES directly into the BLOOD CIRCULATION. These hormones influence the METABOLISM and other functions of cells in the body.
System of herbal medicine practiced in Japan by both herbalists and practitioners of modern medicine. Kampo originated in China and is based on Chinese herbal medicine (MEDICINE, CHINESE TRADITIONAL).
The surgical removal of one or both ovaries.
An aromatase inhibitor that is used in the treatment of advanced BREAST CANCER.
A syndrome characterized by HYPERPIGMENTATION, enlarging pituitary mass, visual defects secondary to compression of the OPTIC CHIASM, and elevated serum ACTH. It is caused by the expansion of an underlying ACTH-SECRETING PITUITARY ADENOMA that grows in the absence of feedback inhibition by adrenal CORTICOSTEROIDS, usually after ADRENALECTOMY.
Cells with the capacity to take up and decarboxylate the amine precursors DIHYDROXYPHENYLALANINE or 5-HYDROXYTRYPTOPHAN. This is a property of endocrine cells of neural and non-neural origin. APUDOMA is a general term collectively applied to tumors associated with APUD cells.
A peptide of 44 amino acids in most species that stimulates the release and synthesis of GROWTH HORMONE. GHRF (or GRF) is synthesized by neurons in the ARCUATE NUCLEUS of the HYPOTHALAMUS. After being released into the pituitary portal circulation, GHRF stimulates GH release by the SOMATOTROPHS in the PITUITARY GLAND.
A melanocortin receptor subtype found primarily in MELANOCYTES. It shows specificity for ALPHA-MSH and ADRENOCORTICOTROPIC HORMONE. Loss of function mutations of the type 1 melanocortin receptor account for the majority of red hair and fair skin recessive traits in human.
The means of moving persons, animals, goods, or materials from one place to another.
An adrenal microsomal cytochrome P450 enzyme that catalyzes the 21-hydroxylation of steroids in the presence of molecular oxygen and NADPH-FERRIHEMOPROTEIN REDUCTASE. This enzyme, encoded by CYP21 gene, converts progesterones to precursors of adrenal steroid hormones (CORTICOSTERONE; HYDROCORTISONE). Defects in CYP21 cause congenital adrenal hyperplasia (ADRENAL HYPERPLASIA, CONGENITAL).
The increase in a measurable parameter of a PHYSIOLOGICAL PROCESS, including cellular, microbial, and plant; immunological, cardiovascular, respiratory, reproductive, urinary, digestive, neural, musculoskeletal, ocular, and skin physiological processes; or METABOLIC PROCESS, including enzymatic and other pharmacological processes, by a drug or other chemical.
Genetically identical individuals developed from brother and sister matings which have been carried out for twenty or more generations or by parent x offspring matings carried out with certain restrictions. This also includes animals with a long history of closed colony breeding.
A strain of albino rat developed at the Wistar Institute that has spread widely at other institutions. This has markedly diluted the original strain.
Cells propagated in vitro in special media conducive to their growth. Cultured cells are used to study developmental, morphologic, metabolic, physiologic, and genetic processes, among others.
A benign neoplasm of the ADRENAL CORTEX. It is characterized by a well-defined nodular lesion, usually less than 2.5 cm. Most adrenocortical adenomas are nonfunctional. The functional ones are yellow and contain LIPIDS. Depending on the cell type or cortical zone involved, they may produce ALDOSTERONE; HYDROCORTISONE; DEHYDROEPIANDROSTERONE; and/or ANDROSTENEDIONE.
A mitochondrial cytochrome P450 enzyme that catalyzes the side-chain cleavage of C27 cholesterol to C21 pregnenolone in the presence of molecular oxygen and NADPH-FERRIHEMOPROTEIN REDUCTASE. This enzyme, encoded by CYP11A1 gene, catalyzes the breakage between C20 and C22 which is the initial and rate-limiting step in the biosynthesis of various gonadal and adrenal steroid hormones.
Peptide hormones produced by NEURONS of various regions in the HYPOTHALAMUS. They are released into the pituitary portal circulation to stimulate or inhibit PITUITARY GLAND functions. VASOPRESSIN and OXYTOCIN, though produced in the hypothalamus, are not included here for they are transported down the AXONS to the POSTERIOR LOBE OF PITUITARY before being released into the portal circulation.
A 14-amino acid peptide named for its ability to inhibit pituitary GROWTH HORMONE release, also called somatotropin release-inhibiting factor. It is expressed in the central and peripheral nervous systems, the gut, and other organs. SRIF can also inhibit the release of THYROID-STIMULATING HORMONE; PROLACTIN; INSULIN; and GLUCAGON besides acting as a neurotransmitter and neuromodulator. In a number of species including humans, there is an additional form of somatostatin, SRIF-28 with a 14-amino acid extension at the N-terminal.
Hormones synthesized from amino acids. They are distinguished from INTERCELLULAR SIGNALING PEPTIDES AND PROTEINS in that their actions are systemic.
A cyclic nucleotide derivative that mimics the action of endogenous CYCLIC AMP and is capable of permeating the cell membrane. It has vasodilator properties and is used as a cardiac stimulant. (From Merck Index, 11th ed)
A 21-carbon steroid, derived from CHOLESTEROL and found in steroid hormone-producing tissues. Pregnenolone is the precursor to GONADAL STEROID HORMONES and the adrenal CORTICOSTEROIDS.
The measurement of an organ in volume, mass, or heaviness.
The decrease in a measurable parameter of a PHYSIOLOGICAL PROCESS, including cellular, microbial, and plant; immunological, cardiovascular, respiratory, reproductive, urinary, digestive, neural, musculoskeletal, ocular, and skin physiological processes; or METABOLIC PROCESS, including enzymatic and other pharmacological processes, by a drug or other chemical.
A general class of ortho-dihydroxyphenylalkylamines derived from tyrosine.
A 51-amino acid pancreatic hormone that plays a major role in the regulation of glucose metabolism, directly by suppressing endogenous glucose production (GLYCOGENOLYSIS; GLUCONEOGENESIS) and indirectly by suppressing GLUCAGON secretion and LIPOLYSIS. Native insulin is a globular protein comprised of a zinc-coordinated hexamer. Each insulin monomer containing two chains, A (21 residues) and B (30 residues), linked by two disulfide bonds. Insulin is used as a drug to control insulin-dependent diabetes mellitus (DIABETES MELLITUS, TYPE 1).
A glycoprotein that causes regression of MULLERIAN DUCTS. It is produced by SERTOLI CELLS of the TESTES. In the absence of this hormone, the Mullerian ducts develop into structures of the female reproductive tract. In males, defects of this hormone result in persistent Mullerian duct, a form of MALE PSEUDOHERMAPHRODITISM.
Hormones produced by the GONADS, including both steroid and peptide hormones. The major steroid hormones include ESTRADIOL and PROGESTERONE from the OVARY, and TESTOSTERONE from the TESTIS. The major peptide hormones include ACTIVINS and INHIBINS.
High affinity receptors for THYROID HORMONES, especially TRIIODOTHYRONINE. These receptors are usually found in the nucleus where they regulate DNA transcription. They are encoded by the THRB gene (also known as NR1A2, THRB1, or ERBA2 gene) as several isoforms produced by alternative splicing. Mutations in the THRB gene cause THYROID HORMONE RESISTANCE SYNDROME.
Precursor of epinephrine that is secreted by the adrenal medulla and is a widespread central and autonomic neurotransmitter. Norepinephrine is the principal transmitter of most postganglionic sympathetic fibers and of the diffuse projection system in the brain arising from the locus ceruleus. It is also found in plants and is used pharmacologically as a sympathomimetic.
A 29-amino acid pancreatic peptide derived from proglucagon which is also the precursor of intestinal GLUCAGON-LIKE PEPTIDES. Glucagon is secreted by PANCREATIC ALPHA CELLS and plays an important role in regulation of BLOOD GLUCOSE concentration, ketone metabolism, and several other biochemical and physiological processes. (From Gilman et al., Goodman and Gilman's The Pharmacological Basis of Therapeutics, 9th ed, p1511)
A highly specific (Leu-Leu) endopeptidase that generates ANGIOTENSIN I from its precursor ANGIOTENSINOGEN, leading to a cascade of reactions which elevate BLOOD PRESSURE and increase sodium retention by the kidney in the RENIN-ANGIOTENSIN SYSTEM. The enzyme was formerly listed as EC 3.4.99.19.
The rate dynamics in chemical or physical systems.
Specific molecular sites or proteins on or in cells to which VASOPRESSINS bind or interact in order to modify the function of the cells. Two types of vasopressin receptor exist, the V1 receptor in the vascular smooth muscle and the V2 receptor in the kidneys. The V1 receptor can be subdivided into V1a and V1b (formerly V3) receptors.
The age of the conceptus, beginning from the time of FERTILIZATION. In clinical obstetrics, the gestational age is often estimated as the time from the last day of the last MENSTRUATION which is about 2 weeks before OVULATION and fertilization.
Injections into the cerebral ventricles.
Any of various animals that constitute the family Suidae and comprise stout-bodied, short-legged omnivorous mammals with thick skin, usually covered with coarse bristles, a rather long mobile snout, and small tail. Included are the genera Babyrousa, Phacochoerus (wart hogs), and Sus, the latter containing the domestic pig (see SUS SCROFA).
The unborn young of a viviparous mammal, in the postembryonic period, after the major structures have been outlined. In humans, the unborn young from the end of the eighth week after CONCEPTION until BIRTH, as distinguished from the earlier EMBRYO, MAMMALIAN.
Compounds that interact with ANDROGEN RECEPTORS in target tissues to bring about the effects similar to those of TESTOSTERONE. Depending on the target tissues, androgenic effects can be on SEX DIFFERENTIATION; male reproductive organs, SPERMATOGENESIS; secondary male SEX CHARACTERISTICS; LIBIDO; development of muscle mass, strength, and power.
'Housing, Animal' refers to the physical structure or environment designed and constructed to provide shelter, protection, and specific living conditions for various domestic or captive animals, meeting their biological and behavioral needs while ensuring their welfare and well-being.
The restriction of the MOVEMENT of whole or part of the body by physical means (RESTRAINT, PHYSICAL) or chemically by ANALGESIA, or the use of TRANQUILIZING AGENTS or NEUROMUSCULAR NONDEPOLARIZING AGENTS. It includes experimental protocols used to evaluate the physiologic effects of immobility.
Antibiotic substance isolated from streptomycin-producing strains of Streptomyces griseus. It acts by inhibiting elongation during protein synthesis.
The mass or quantity of heaviness of an individual. It is expressed by units of pounds or kilograms.
HORMONES secreted by the gastrointestinal mucosa that affect the timing or the quality of secretion of digestive enzymes, and regulate the motor activity of the digestive system organs.
A peptide hormone that lowers calcium concentration in the blood. In humans, it is released by thyroid cells and acts to decrease the formation and absorptive activity of osteoclasts. Its role in regulating plasma calcium is much greater in children and in certain diseases than in normal adults.
Any of the processes by which nuclear, cytoplasmic, or intercellular factors influence the differential control (induction or repression) of gene action at the level of transcription or translation.
Descriptions of specific amino acid, carbohydrate, or nucleotide sequences which have appeared in the published literature and/or are deposited in and maintained by databanks such as GENBANK, European Molecular Biology Laboratory (EMBL), National Biomedical Research Foundation (NBRF), or other sequence repositories.
Histochemical localization of immunoreactive substances using labeled antibodies as reagents.
The range or frequency distribution of a measurement in a population (of organisms, organs or things) that has not been selected for the presence of disease or abnormality.
A process involving chance used in therapeutic trials or other research endeavor for allocating experimental subjects, human or animal, between treatment and control groups, or among treatment groups. It may also apply to experiments on inanimate objects.
A tripeptide that stimulates the release of THYROTROPIN and PROLACTIN. It is synthesized by the neurons in the PARAVENTRICULAR NUCLEUS of the HYPOTHALAMUS. After being released into the pituitary portal circulation, TRH (was called TRF) stimulates the release of TSH and PRL from the ANTERIOR PITUITARY GLAND.
An increase in the number of cells in a tissue or organ without tumor formation. It differs from HYPERTROPHY, which is an increase in bulk without an increase in the number of cells.
Naturally occurring or experimentally induced animal diseases with pathological processes sufficiently similar to those of human diseases. They are used as study models for human diseases.
An octapeptide that is a potent but labile vasoconstrictor. It is produced from angiotensin I after the removal of two amino acids at the C-terminal by ANGIOTENSIN CONVERTING ENZYME. The amino acid in position 5 varies in different species. To block VASOCONSTRICTION and HYPERTENSION effect of angiotensin II, patients are often treated with ACE INHIBITORS or with ANGIOTENSIN II TYPE 1 RECEPTOR BLOCKERS.
High affinity receptors for THYROID HORMONES, especially TRIIODOTHYRONINE. These receptors are usually found in the nucleus where they regulate DNA transcription. They are encoded by the THRA gene (also known as NR1A1, THRA1, ERBA or ERBA1 gene) as several isoforms produced by alternative splicing.
The sequence of PURINES and PYRIMIDINES in nucleic acids and polynucleotides. It is also called nucleotide sequence.
The alpha chain of pituitary glycoprotein hormones (THYROTROPIN; FOLLICLE STIMULATING HORMONE; LUTEINIZING HORMONE) and the placental CHORIONIC GONADOTROPIN. Within a species, the alpha subunits of these four hormones are identical; the distinct functional characteristics of these glycoprotein hormones are determined by the unique beta subunits. Both subunits, the non-covalently bound heterodimers, are required for full biologic activity.
The domestic dog, Canis familiaris, comprising about 400 breeds, of the carnivore family CANIDAE. They are worldwide in distribution and live in association with people. (Walker's Mammals of the World, 5th ed, p1065)
Hormones secreted by insects. They influence their growth and development. Also synthetic substances that act like insect hormones.
Glucose in blood.
The process of bearing developing young (EMBRYOS or FETUSES) in utero in non-human mammals, beginning from FERTILIZATION to BIRTH.
Cellular DNA-binding proteins encoded by the c-fos genes (GENES, FOS). They are involved in growth-related transcriptional control. c-fos combines with c-jun (PROTO-ONCOGENE PROTEINS C-JUN) to form a c-fos/c-jun heterodimer (TRANSCRIPTION FACTOR AP-1) that binds to the TRE (TPA-responsive element) in promoters of certain genes.
Those characteristics that distinguish one SEX from the other. The primary sex characteristics are the OVARIES and TESTES and their related hormones. Secondary sex characteristics are those which are masculine or feminine but not directly related to reproduction.
Chemical substances which inhibit the function of the endocrine glands, the biosynthesis of their secreted hormones, or the action of hormones upon their specific sites.
Injections made into a vein for therapeutic or experimental purposes.

Cortisol in fetal fluids and the fetal adrenal at parturition in the tammar wallaby (Macropus eugenii). (1/3421)

Glucocorticoid hormones may play a critical role in initiating parturition in tammar wallabies. In this study, we investigated the concentration of cortisol in fetal fluids and cortisol production by fetal adrenals over the last 3 days of the 26-day pregnancy and within 24 h postpartum. The fetal adrenals almost doubled in size between Days 24 and 26 of pregnancy, and their cortisol content increased over 10-fold during this period, from 10 pg to over 100 pg per adrenal pair. After birth, neonatal adrenals continued to grow, but cortisol content fell dramatically to 20 pg. The prepartum increase in adrenal cortisol was reflected by a substantial rise in cortisol concentrations in yolk sac fluid, allantoic fluid, and fetal blood, which were below 10 ng/ml on Day 24 and rose to over 40 ng/ml by Day 26. Cortisol concentrations in neonatal blood decreased postpartum, mirroring decreased cortisol content in neonatal adrenals. Cortisol production by the fetal adrenal was stimulated in vitro by ACTH and prostaglandin E2, suggesting that the in vivo increase may be stimulated by release of ACTH from the fetal hypothalamic-pituitary axis and prostaglandin E2 from the placenta. These results indicate that increasing cortisol production by the fetal adrenal is a characteristic of late pregnancy in the tammar wallaby and support the suggestion that fetal cortisol may trigger the initiation of parturition in this marsupial species.  (+info)

Delay of preterm delivery in sheep by omega-3 long-chain polyunsaturates. (2/3421)

A positive correlation has been shown between dietary intake of long-chain omega-3 fatty acids in late pregnancy and gestation length in pregnant women and experimental animals. To determine whether omega-3 fatty acids have an effect on preterm labor in sheep, a fish oil concentrate emulsion was continuously infused to six pregnant ewes from 124 days gestational age. At 125 days, betamethasone was administered to the fetus to produce preterm labor. Both the onset of labor and the time of delivery were delayed by the fish oil emulsion. Two of the omega-3-infused ewes reverted from contractions to nonlabor, an effect never previously observed for experimental glucocorticoid-induced preterm labor in sheep. Maternal plasma estradiol and maternal and fetal prostaglandin E2 rose in control ewes but not in those infused with omega-3 fatty acid. The ability of omega-3 fatty acids to delay premature delivery in sheep indicates their possible use as tocolytics in humans. Premature labor is the major cause of neonatal death and long-term disability, and these studies present information that may lead to a novel therapeutic regimen for the prevention of preterm delivery in human pregnancy.  (+info)

On the meaning of low-dose ACTH(1-24) tests to assess functionality of the hypothalamic-pituitary-adrenal axis. (3/3421)

To analyse further the ACTH(1-24) low-dose test, which is of clinical interest, we have examined the dose-response relationship between plasma ACTH(1-24) and cortisol concentrations after i.v. administration of increasing doses (1, 5 or 250 microg) of ACTH(1-24) as a bolus. In addition, we have measured plasma ACTH(1-39) and cortisol levels after an insulin tolerance test (ITT). Although there was a dose response relationship between plasma ACTH(1-24) immunoreactivity and the dose injected, cortisol peaks were comparable, but lower than those reached after an ITT. Under these experimental conditions, an increase in plasma ACTH as low as 13 pmol/l (i.e. the increase obtained with the 1 microg dose) induced a near maximal cortisol response. Following injection of 1 microg ACTH(1-24), peak ACTH values were short lasting, similar to physiological daily bursts. After injection of 5 microg ACTH(1-24), plasma ACTH concentrations were higher than those reached during an ITT, but clearly shorter lasting. Injection of 250 microg ACTH(1-24) induced strikingly supraphysiological levels of plasma ACTH. We conclude that neither regular nor low-dose ACTH tests can fully reproduce the ITT. Our observations strongly suggest that the low-dose ACTH(1-24) test (1 microg) can be useful to estimate the adrenal sensitivity under basal, physiological conditions.  (+info)

The treatment of insulin resistance does not improve adrenal cytochrome P450c17alpha enzyme dysregulation in polycystic ovary syndrome. (4/3421)

OBJECTIVE: To determine whether metformin. when given to non-diabetic women with polycystic ovary syndrome (PCOS), results in a reduction of insulin resistance and hyperinsulinemia while body weight is maintained. Also we aimed to see whether the reduction in insulin levels attenuates the activity of adrenal P450c17alpha enzyme in patients with PCOS. DESIGN: We investigated the 17-hydroxyprogesterone (17-OHP) and androstenedione responses to ACTH, insulin responses to an oral glucose tolerance test (OGTT) and glucose disposal rate in an insulin tolerance test before and after metformin therapy (500 mg, orally, twice daily, for 12 weeks). METHODS: The presence of hyperinsulinemia in 15 women with PCOS was demonstrated by an OGTT and results were compared with those of 10 healthy women. Insulin sensitivity was measured by the rate of endogenous glucose disposal after i.v. bolus injection of insulin. 17-OHP and androstenedione responses to ACTH were measured in all the women with PCOS and the normal women. RESULTS: Women with PCOS were hyperinsulinemic (102.0+/-13.0 (S.E.M.) VS 46.2+/-4.4 pmol/l) and hyperandrogenemic (free testosterone 15.3+/-1.7 vs 7.9+/-0.6 nmol/l; androstenedione 11.8+/-0.8 vs 8.2+/-0.6 nmol/l) and more hirsute (modified Ferriman-Gallwey score, 17.7+/-1.6 vs 3.0+/-0.3) than healthy women. In addition, women with PCOS had higher 17-OHP and androstenedione responses to ACTH when compared with healthy women. Metformin therapy resulted in some improvement in insulin sensitivity and reduced the basal and post-glucose load insulin levels. But 17-OHP and androstenedione responses to ACTH were unaltered in response to metformin. CONCLUSIONS: PCOS is characterized by hyperactivity of the adrenal P450c17alpha enzyme and insulin resistance. It seems that there is no direct relationship between insulin resistance and adrenal P450c17alpha enzyme dysregulation.  (+info)

Primary hypoadrenocorticism in a dog receiving glucocorticoid supplementation. (5/3421)

A 5-year-old, spayed, female husky-Labrador retriever cross was diagnosed with primary hypoadrenocorticism, an uncommon endocrine disorder caused by a deficiency of glucocorticoid and mineralocorticoid hormones. Subtle clinical signs and previous treatment with exogenous glucocorticoid drugs required an adrenocorticotropic hormone stimulation test to confirm the diagnosis.  (+info)

Central administration of rat IL-6 induces HPA activation and fever but not sickness behavior in rats. (6/3421)

Interleukin (IL)-6 has been proposed to mediate several sickness responses, including brain-mediated neuroendocrine, temperature, and behavioral changes. However, the exact mechanisms and sites of action of IL-6 are still poorly understood. In the present study, we describe the effects of central administration of species-homologous recombinant rat IL-6 (rrIL-6) on the induction of hypothalamic-pituitary-adrenal (HPA) activity, fever, social investigatory behavior, and immobility. After intracerebroventricular administration of rrIL-6 (50 or 100 ng/rat), rats demonstrated HPA and febrile responses. In contrast, rrIL-6 alone did not induce changes in social investigatory and locomotor behavior at doses of up to 400 ng/rat. Coadministration of rrIL-6 (100 ng/rat) and rrIL-1beta (40 ng/rat), which alone did not affect the behavioral responses, reduced social investigatory behavior and increased the duration of immobility. Compared with rhIL-6, intracerebroventricular administration of rrIL-6 (100 ng/rat) induced higher HPA responses and early-phase febrile responses. This is consistent with a higher potency of rrIL-6, compared with rhIL-6, in the murine B9 bioassay. We conclude that species-homologous rrIL-6 alone can act in the brain to induce HPA and febrile responses, whereas it only reduces social investigatory behavior and locomotor activity in the presence of IL-1beta.  (+info)

Suppression of the secretion of luteinizing hormone due to isolation/restraint stress in gonadectomised rams and ewes is influenced by sex steroids. (7/3421)

In this study we used an isolation/restraint stress to test the hypothesis that stress will affect the secretion of LH differently in gonadectomised rams and ewes treated with different combinations of sex steroids. Romney Marsh sheep were gonadectomised two weeks prior to these experiments. In the first experiment male and female sheep were treated with vehicle or different sex steroids for 7 days prior to the application of the isolation/restraint stress. Male sheep received either i.m. oil (control rams) or 6 mg testosterone propionate injections every 12 h. Female sheep were given empty s.c. implants (control ewes), or 2x1 cm s.c. implants containing oestradiol, or an intravaginal controlled internal drug release device containing 0.3 g progesterone, or the combination of oestradiol and progesterone. There were four animals in each group. On the day of application of the isolation/restraint stress, blood samples were collected every 10 min for 16 h for the subsequent measurement of plasma LH and cortisol concentrations. After 8 h the stress was applied for 4 h. Two weeks later, blood samples were collected for a further 16 h from the control rams and ewes, but on this day no stress was imposed. In the second experiment, separate control gonadectomised rams and ewes (n=4/group) were studied for 7 h on 3 consecutive days, when separate treatments were applied. On day 1, the animals received no treatment; on day 2, isolation/restraint stress was applied after 3 h; and on day 3, an i. v. injection of 2 microg/kg ACTH1-24 was given after 3 h. On each day, blood samples were collected every 10 min and the LH response to the i.v. injection of 500 ng GnRH administered after 5 h of sampling was measured. In Experiment 1, the secretion of LH was suppressed during isolation/restraint in all groups but the parameters of LH secretion (LH pulse frequency and amplitude) that were affected varied between groups. In control rams, LH pulse amplitude, and not frequency, was decreased during isolation/restraint whereas in rams treated with testosterone propionate the stressor reduced pulse frequency and not amplitude. In control ewes, isolation/restraint decreased LH pulse frequency but not amplitude. Isolation/restraint reduced both LH pulse frequency and amplitude in ewes treated with oestradiol, LH pulse frequency in ewes treated with progesterone and only LH pulse amplitude in ewes treated with both oestradiol and progesterone. There was no change in LH secretion during the day of no stress. Plasma concentrations of cortisol were higher during isolation/restraint than on the day of no stress. On the day of isolation/restraint maximal concentrations of cortisol were observed during the application of the stressor but there were no differences between groups in the magnitude of this response. In Experiment 2, isolation/restraint reduced the LH response to GnRH in rams but not ewes and ACTH reduced the LH response to GnRH both in rams and ewes. Our results show that the mechanism(s) by which isolation/restraint stress suppresses LH secretion in sheep is influenced by sex steroids. The predominance of particular sex steroids in the circulation may affect the extent to which stress inhibits the secretion of GnRH from the hypothalamus and/or the responsiveness of the pituitary gland to the actions of GnRH. There are also differences between the sexes in the effects of stress on LH secretion that are independent of the sex steroids.  (+info)

Tests of adrenal insufficiency. (8/3421)

AIM: In suspected adrenal insufficiency, the ideal test for assessing the hypothalamo-pituitary-adrenal axis is controversial. Therefore, three tests were compared in patients presenting with symptoms suggestive of adrenal insufficiency. METHOD: Responses to the standard short Synacthen test (SSST), the low dose Synacthen test (LDST), and the 08:00 hour serum cortisol concentration were measured in 32 patients. A normal response to the synacthen test was defined as a peak serum cortisol of >/= 500 nmol/l and/or incremental concentration of >/= 200 nmol/l. The sensitivity and specificity of the 08:00 hour serum cortisol concentration compared with other tests was calculated. RESULTS: Three patients had neither an adequate peak nor increment after the SSST and LDST. All had a serum 08:00 hour cortisol concentration of < 200 nmol/l. Eight patients had abnormal responses by both criteria to the LDST but had normal responses to the SSST. Three reported amelioration of their symptoms on hydrocortisone replacement. Twenty one patients had a normal response to both tests (of these, 14 achieved adequate peak and increment after both tests and seven did not have an adequate peak after the LDST but had a normal increment). The lowest 08:00 hour serum cortisol concentration above which patients achieved normal responses to both the LDST and SSST was 500 nmol/l. At this cut off value (compared with the LDST), the serum 08:00 hour cortisol concentration had a sensitivity of 100% but specificity was only 33%. CONCLUSION: The LDST revealed mild degrees of adrenal insufficiency not detected by the SSST. The value of a single 08:00 hour serum cortisol concentration is limited.  (+info)

Adrenocorticotropic Hormone (ACTH) is a hormone produced and released by the anterior pituitary gland, a small endocrine gland located at the base of the brain. ACTH plays a crucial role in the regulation of the body's stress response and has significant effects on various physiological processes.

The primary function of ACTH is to stimulate the adrenal glands, which are triangular-shaped glands situated on top of the kidneys. The adrenal glands consist of two parts: the outer cortex and the inner medulla. ACTH specifically targets the adrenal cortex, where it binds to specific receptors and initiates a series of biochemical reactions leading to the production and release of steroid hormones, primarily cortisol (a glucocorticoid) and aldosterone (a mineralocorticoid).

Cortisol is involved in various metabolic processes, such as regulating blood sugar levels, modulating the immune response, and helping the body respond to stress. Aldosterone plays a vital role in maintaining electrolyte and fluid balance by promoting sodium reabsorption and potassium excretion in the kidneys.

ACTH release is controlled by the hypothalamus, another part of the brain, which produces corticotropin-releasing hormone (CRH). CRH stimulates the anterior pituitary gland to secrete ACTH, which in turn triggers cortisol production in the adrenal glands. This complex feedback system helps maintain homeostasis and ensures that appropriate amounts of cortisol are released in response to various physiological and psychological stressors.

Disorders related to ACTH can lead to hormonal imbalances, resulting in conditions such as Cushing's syndrome (excessive cortisol production) or Addison's disease (insufficient cortisol production). Proper diagnosis and management of these disorders typically involve assessing the function of the hypothalamic-pituitary-adrenal axis and addressing any underlying issues affecting ACTH secretion.

Hydrocortisone is a synthetic glucocorticoid, which is a class of steroid hormones. It is identical to the naturally occurring cortisol, a hormone produced by the adrenal gland that helps regulate metabolism and helps your body respond to stress. Hydrocortisone has anti-inflammatory effects and is used to treat various inflammatory conditions such as allergies, skin disorders, and autoimmune diseases. It works by suppressing the immune system's response to reduce swelling, redness, itching, and other symptoms caused by inflammation.

Hydrocortisone is available in different forms, including oral tablets, topical creams, lotions, gels, and ointments, as well as injectable solutions. The specific use and dosage depend on the condition being treated and the individual patient's medical history and current health status.

As with any medication, hydrocortisone can have side effects, especially when used in high doses or for extended periods. Common side effects include increased appetite, weight gain, mood changes, insomnia, and skin thinning. Long-term use of hydrocortisone may also increase the risk of developing osteoporosis, diabetes, cataracts, and other health problems. Therefore, it is essential to follow your healthcare provider's instructions carefully when using this medication.

The adrenal glands are a pair of endocrine glands that are located on top of the kidneys. Each gland has two parts: the outer cortex and the inner medulla. The adrenal cortex produces hormones such as cortisol, aldosterone, and androgens, which regulate metabolism, blood pressure, and other vital functions. The adrenal medulla produces catecholamines, including epinephrine (adrenaline) and norepinephrine (noradrenaline), which help the body respond to stress by increasing heart rate, blood pressure, and alertness.

The pituitary-adrenal system, also known as the hypothalamic-pituitary-adrenal (HPA) axis, is a complex set of interactions between the hypothalamus, the pituitary gland, and the adrenal glands. This system plays a crucial role in the body's response to stress through the release of hormones that regulate various physiological processes.

The hypothalamus, located within the brain, receives information from the nervous system about the internal and external environment and responds by releasing corticotropin-releasing hormone (CRH) and vasopressin. These hormones then travel to the anterior pituitary gland, where they stimulate the release of adrenocorticotropic hormone (ACTH).

ACTH is transported through the bloodstream to the adrenal glands, which are located on top of the kidneys. The adrenal glands consist of two parts: the outer cortex and the inner medulla. ACTH specifically targets the adrenal cortex, causing it to release cortisol and other glucocorticoids, as well as androgens such as dehydroepiandrosterone (DHEA).

Cortisol has numerous effects on metabolism, immune function, and cardiovascular regulation. It helps regulate blood sugar levels, suppresses the immune system, and aids in the breakdown of fats, proteins, and carbohydrates to provide energy during stressful situations. DHEA can be converted into male and female sex hormones (androgens and estrogens) in various tissues throughout the body.

The pituitary-adrenal system is tightly regulated through negative feedback mechanisms. High levels of cortisol, for example, inhibit the release of CRH and ACTH from the hypothalamus and pituitary gland, respectively, thereby limiting further cortisol production. Dysregulation of this system has been implicated in several medical conditions, including Cushing's syndrome (overproduction of cortisol) and Addison's disease (underproduction of cortisol).

Hormones are defined as chemical messengers that are produced by endocrine glands or specialized cells and are transported through the bloodstream to tissues and organs, where they elicit specific responses. They play crucial roles in regulating various physiological processes such as growth, development, metabolism, reproduction, and mood. Examples of hormones include insulin, estrogen, testosterone, adrenaline, and thyroxine.

The Hypothalamo-Hypophyseal system, also known as the hypothalamic-pituitary system, is a crucial part of the endocrine system that regulates many bodily functions. It consists of two main components: the hypothalamus and the pituitary gland.

The hypothalamus is a region in the brain that receives information from various parts of the body and integrates them to regulate vital functions such as body temperature, hunger, thirst, sleep, and emotional behavior. It also produces and releases neurohormones that control the secretion of hormones from the pituitary gland.

The pituitary gland is a small gland located at the base of the brain, just below the hypothalamus. It consists of two parts: the anterior pituitary (also called adenohypophysis) and the posterior pituitary (also called neurohypophysis). The anterior pituitary produces and releases several hormones that regulate various bodily functions such as growth, metabolism, reproduction, and stress response. The posterior pituitary stores and releases hormones produced by the hypothalamus, including antidiuretic hormone (ADH) and oxytocin.

The hypothalamo-hypophyseal system works together to maintain homeostasis in the body by regulating various physiological processes through hormonal signaling. Dysfunction of this system can lead to several endocrine disorders, such as diabetes insipidus, pituitary tumors, and hypothalamic-pituitary axis disorders.

Corticotropin-Releasing Hormone (CRH) is a hormone that is produced and released by the hypothalamus, a small gland located in the brain. CRH plays a critical role in the body's stress response system.

When the body experiences stress, the hypothalamus releases CRH, which then travels to the pituitary gland, another small gland located at the base of the brain. Once there, CRH stimulates the release of adrenocorticotropic hormone (ACTH) from the pituitary gland.

ACTH then travels through the bloodstream to the adrenal glands, which are located on top of the kidneys. ACTH stimulates the adrenal glands to produce and release cortisol, a hormone that helps the body respond to stress by regulating metabolism, immune function, and blood pressure, among other things.

Overall, CRH is an important part of the hypothalamic-pituitary-adrenal (HPA) axis, which regulates many bodily functions related to stress response, mood, and cognition. Dysregulation of the HPA axis and abnormal levels of CRH have been implicated in various psychiatric and medical conditions, including depression, anxiety disorders, post-traumatic stress disorder (PTSD), and Cushing's syndrome.

Infantile spasms, also known as West syndrome, is a rare but serious type of epilepsy that affects infants typically between 4-8 months of age. The spasms are characterized by sudden, brief, and frequent muscle jerks or contractions, often involving the neck, trunk, and arms. These spasms usually occur in clusters and may cause the infant to bend forward or stretch out. Infantile spasms can be a symptom of various underlying neurological conditions and are often associated with developmental delays and regression. Early recognition and treatment are crucial for improving outcomes.

Corticosterone is a hormone produced by the adrenal gland in many animals, including humans. It is a type of glucocorticoid steroid hormone that plays an important role in the body's response to stress, immune function, metabolism, and regulation of inflammation. Corticosterone helps to regulate the balance of sodium and potassium in the body and also plays a role in the development and functioning of the nervous system. It is the primary glucocorticoid hormone in rodents, while cortisol is the primary glucocorticoid hormone in humans and other primates.

The adrenal cortex is the outer portion of the adrenal gland, which is located on top of the kidneys. It plays a crucial role in producing hormones that are essential for various bodily functions. The adrenal cortex is divided into three zones:

1. Zona glomerulosa: This outermost zone produces mineralocorticoids, primarily aldosterone. Aldosterone helps regulate sodium and potassium balance and thus influences blood pressure by controlling the amount of fluid in the body.
2. Zona fasciculata: The middle layer is responsible for producing glucocorticoids, with cortisol being the most important one. Cortisol regulates metabolism, helps manage stress responses, and has anti-inflammatory properties. It also plays a role in blood sugar regulation and maintaining the body's response to injury and illness.
3. Zona reticularis: The innermost zone produces androgens, primarily dehydroepiandrosterone (DHEA) and its sulfate form (DHEAS). These androgens are weak compared to those produced by the gonads (ovaries or testes), but they can be converted into more potent androgens or estrogens in peripheral tissues.

Disorders related to the adrenal cortex can lead to hormonal imbalances, affecting various bodily functions. Examples include Addison's disease (insufficient adrenal cortical hormone production) and Cushing's syndrome (excessive glucocorticoid levels).

Cushing syndrome is a hormonal disorder that occurs when your body is exposed to high levels of the hormone cortisol for a long time. This can happen due to various reasons such as taking high doses of corticosteroid medications or tumors that produce cortisol or adrenocorticotropic hormone (ACTH).

The symptoms of Cushing syndrome may include:

* Obesity, particularly around the trunk and upper body
* Thinning of the skin, easy bruising, and purple or red stretch marks on the abdomen, thighs, breasts, and arms
* Weakened bones, leading to fractures
* High blood pressure
* High blood sugar
* Mental changes such as depression, anxiety, and irritability
* Increased fatigue and weakness
* Menstrual irregularities in women
* Decreased fertility in men

Cushing syndrome can be diagnosed through various tests, including urine and blood tests to measure cortisol levels, saliva tests, and imaging tests to locate any tumors. Treatment depends on the cause of the condition but may include surgery, radiation therapy, chemotherapy, or adjusting medication dosages.

Adrenal insufficiency is a condition in which the adrenal glands do not produce adequate amounts of certain hormones, primarily cortisol and aldosterone. Cortisol helps regulate metabolism, respond to stress, and suppress inflammation, while aldosterone helps regulate sodium and potassium levels in the body to maintain blood pressure.

Primary adrenal insufficiency, also known as Addison's disease, occurs when there is damage to the adrenal glands themselves, often due to autoimmune disorders, infections, or certain medications. Secondary adrenal insufficiency occurs when the pituitary gland fails to produce enough adrenocorticotropic hormone (ACTH), which stimulates the adrenal glands to produce cortisol.

Symptoms of adrenal insufficiency may include fatigue, weakness, weight loss, decreased appetite, nausea, vomiting, diarrhea, abdominal pain, low blood pressure, dizziness, and darkening of the skin. Treatment typically involves replacing the missing hormones with medications taken orally or by injection.

Corticotropin receptors are a type of cell surface receptor that bind to the hormone corticotropin (also known as adrenocorticotropic hormone or ACTH). These receptors are found in various tissues throughout the body, including the adrenal glands.

There are two main types of corticotropin receptors, known as melanocortin receptor 1 (MC1R) and melanocortin receptor 2 (MC2R). MC2R is the primary receptor for corticotropin in the adrenal glands. When corticotropin binds to this receptor, it stimulates the production and release of steroid hormones, such as cortisol, which help regulate metabolism, immune response, and stress response.

Abnormalities in corticotropin receptors have been implicated in several medical conditions, including certain endocrine disorders and skin pigmentation disorders.

The pituitary gland is a small, endocrine gland located at the base of the brain, in the sella turcica of the sphenoid bone. It is often called the "master gland" because it controls other glands and makes the hormones that trigger many body functions. The pituitary gland measures about 0.5 cm in height and 1 cm in width, and it weighs approximately 0.5 grams.

The pituitary gland is divided into two main parts: the anterior lobe (adenohypophysis) and the posterior lobe (neurohypophysis). The anterior lobe is further divided into three zones: the pars distalis, pars intermedia, and pars tuberalis. Each part of the pituitary gland has distinct functions and produces different hormones.

The anterior pituitary gland produces and releases several important hormones, including:

* Growth hormone (GH), which regulates growth and development in children and helps maintain muscle mass and bone strength in adults.
* Thyroid-stimulating hormone (TSH), which controls the production of thyroid hormones by the thyroid gland.
* Adrenocorticotropic hormone (ACTH), which stimulates the adrenal glands to produce cortisol and other steroid hormones.
* Follicle-stimulating hormone (FSH) and luteinizing hormone (LH), which regulate reproductive function in both males and females.
* Prolactin, which stimulates milk production in pregnant and lactating women.

The posterior pituitary gland stores and releases two hormones that are produced by the hypothalamus:

* Antidiuretic hormone (ADH), which helps regulate water balance in the body by controlling urine production.
* Oxytocin, which stimulates uterine contractions during childbirth and milk release during breastfeeding.

Overall, the pituitary gland plays a critical role in maintaining homeostasis and regulating various bodily functions, including growth, development, metabolism, and reproductive function.

Melanocyte-stimulating hormones (MSH) are a group of peptide hormones that originate from the precursor protein proopiomelanocortin (POMC). They play crucial roles in various physiological processes, including pigmentation, energy balance, and appetite regulation.

There are several types of MSH, but the most well-known ones include α-MSH, β-MSH, and γ-MSH. These hormones bind to melanocortin receptors (MCRs), which are found in various tissues throughout the body. The binding of MSH to MCRs triggers a series of intracellular signaling events that ultimately lead to changes in cell behavior.

In the context of skin physiology, α-MSH and β-MSH bind to melanocortin 1 receptor (MC1R) on melanocytes, which are the cells responsible for producing pigment (melanin). This binding stimulates the production and release of eumelanin, a type of melanin that is brown or black in color. As a result, increased levels of MSH can lead to darkening of the skin, also known as hyperpigmentation.

Apart from their role in pigmentation, MSH hormones have been implicated in several other physiological processes. For instance, α-MSH has been shown to suppress appetite and promote weight loss by binding to melanocortin 4 receptor (MC4R) in the hypothalamus, a region of the brain that regulates energy balance. Additionally, MSH hormones have been implicated in inflammation, immune response, and sexual function.

Overall, melanocyte-stimulating hormones are a diverse group of peptide hormones that play important roles in various physiological processes, including pigmentation, energy balance, and appetite regulation.

Cosyntropin is a synthetic form of adrenocorticotropic hormone (ACTH) that is used in medical testing to assess the function of the adrenal glands. ACTH is a hormone produced and released by the pituitary gland that stimulates the production and release of cortisol, a steroid hormone produced by the adrenal glands.

Cosyntropin is typically administered as an injection, and its effects on cortisol production are measured through blood tests taken at various time points after administration. This test, known as a cosyntropin stimulation test or ACTH stimulation test, can help diagnose conditions that affect the adrenal glands, such as Addison's disease or adrenal insufficiency.

It is important to note that while cosyntropin is a synthetic form of ACTH, it is not identical to the natural hormone and may have slightly different effects on the body. Therefore, it should only be used under the supervision of a healthcare professional.

A melanocortin type 2 receptor (MC2R) is a G protein-coupled receptor that binds melanocortin peptides such as adrenocorticotropic hormone (ACTH). It is primarily expressed in the adrenal gland, specifically in the zona fasciculata of the cortex. Upon activation by ACTH, MC2R stimulates the production and release of steroid hormones, particularly cortisol, through the cAMP signaling pathway. Dysfunction in this receptor can lead to various endocrine disorders such as congenital adrenal hyperplasia and Cushing's disease.

Adrenocortical hyperfunction, also known as Cushing's syndrome, is a condition characterized by the overproduction of cortisol hormone from the adrenal glands. The adrenal glands are located on top of the kidneys and are responsible for producing several essential hormones, including cortisol. Cortisol helps regulate metabolism, blood pressure, and the body's response to stress.

In Adrenocortical hyperfunction, the adrenal glands produce too much cortisol, leading to a range of symptoms such as weight gain, particularly around the trunk and face, thinning of the skin, easy bruising, muscle weakness, mood changes, and high blood pressure. The condition can be caused by several factors, including tumors in the pituitary gland or adrenal glands, long-term use of corticosteroid medications, or genetic disorders that affect the adrenal glands.

Treatment for Adrenocortical hyperfunction depends on the underlying cause of the condition and may include surgery to remove tumors, medication to reduce cortisol production, or radiation therapy. It is essential to diagnose and treat this condition promptly, as long-term exposure to high levels of cortisol can lead to serious health complications such as diabetes, osteoporosis, and heart disease.

Pro-opiomelanocortin (POMC) is a precursor protein that gets cleaved into several biologically active peptides in the body. These peptides include adrenocorticotropic hormone (ACTH), beta-lipotropin, and multiple opioid peptides such as beta-endorphin, met-enkephalin, and leu-enkephalin.

ACTH stimulates the release of cortisol from the adrenal gland, while beta-lipotropin has various metabolic functions. The opioid peptides derived from POMC have pain-relieving (analgesic) and rewarding effects in the brain. Dysregulation of the POMC system has been implicated in several medical conditions, including obesity, addiction, and certain types of hormone deficiencies.

Follicle-Stimulating Hormone (FSH) is a glycoprotein hormone secreted and released by the anterior pituitary gland. In females, it promotes the growth and development of ovarian follicles in the ovary, which ultimately leads to the maturation and release of an egg (ovulation). In males, FSH stimulates the testes to produce sperm. It works in conjunction with luteinizing hormone (LH) to regulate reproductive processes. The secretion of FSH is controlled by the hypothalamic-pituitary-gonadal axis and its release is influenced by the levels of gonadotropin-releasing hormone (GnRH), estrogen, inhibin, and androgens.

Luteinizing Hormone (LH) is a glycoprotein hormone, which is primarily produced and released by the anterior pituitary gland. In women, a surge of LH triggers ovulation, the release of an egg from the ovaries during the menstrual cycle. During pregnancy, LH stimulates the corpus luteum to produce progesterone. In men, LH stimulates the testes to produce testosterone. It plays a crucial role in sexual development, reproduction, and maintaining the reproductive system.

Pituitary hormones are chemical messengers produced and released by the pituitary gland, a small endocrine gland located at the base of the brain. The pituitary gland is often referred to as the "master gland" because it controls several other endocrine glands and regulates various bodily functions.

There are two main types of pituitary hormones: anterior pituitary hormones and posterior pituitary hormones, which are produced in different parts of the pituitary gland and have distinct functions.

Anterior pituitary hormones include:

1. Growth hormone (GH): regulates growth and metabolism.
2. Thyroid-stimulating hormone (TSH): stimulates the thyroid gland to produce thyroid hormones.
3. Adrenocorticotropic hormone (ACTH): stimulates the adrenal glands to produce cortisol and other steroid hormones.
4. Follicle-stimulating hormone (FSH) and luteinizing hormone (LH): regulate reproductive function in both males and females.
5. Prolactin: stimulates milk production in lactating women.
6. Melanocyte-stimulating hormone (MSH): regulates skin pigmentation and appetite.

Posterior pituitary hormones include:

1. Oxytocin: stimulates uterine contractions during childbirth and milk ejection during lactation.
2. Vasopressin (antidiuretic hormone, ADH): regulates water balance in the body by controlling urine production in the kidneys.

Overall, pituitary hormones play crucial roles in regulating growth, development, metabolism, reproductive function, and various other bodily functions. Abnormalities in pituitary hormone levels can lead to a range of medical conditions, such as dwarfism, acromegaly, Cushing's disease, infertility, and diabetes insipidus.

Ectopic ACTH syndrome is a medical condition characterized by the excessive production of adrenocorticotropic hormone (ACTH) from a source outside of the pituitary gland, typically from a tumor in another part of the body. The most common sources of ectopic ACTH are small-cell lung carcinomas, but it can also occur with other types of tumors such as thymic carcinoids, pancreatic islet cell tumors, and bronchial carcinoids.

The excessive production of ACTH leads to an overproduction of cortisol from the adrenal glands, resulting in a constellation of symptoms known as Cushing's syndrome. These symptoms can include weight gain, muscle weakness, thinning of the skin, easy bruising, mood changes, and high blood pressure, among others.

Ectopic ACTH syndrome is typically more severe than pituitary-dependent Cushing's syndrome, and it may be more difficult to diagnose and treat due to the underlying tumor causing the excessive ACTH production. Treatment usually involves removing the tumor or controlling its growth, as well as managing the symptoms of Cushing's syndrome with medications that block cortisol production or action.

Pituitary neoplasms refer to abnormal growths or tumors in the pituitary gland, a small endocrine gland located at the base of the brain. These neoplasms can be benign (non-cancerous) or malignant (cancerous), with most being benign. They can vary in size and may cause various symptoms depending on their location, size, and hormonal activity.

Pituitary neoplasms can produce and secrete excess hormones, leading to a variety of endocrine disorders such as Cushing's disease (caused by excessive ACTH production), acromegaly (caused by excessive GH production), or prolactinoma (caused by excessive PRL production). They can also cause local compression symptoms due to their size, leading to headaches, vision problems, and cranial nerve palsies.

The exact causes of pituitary neoplasms are not fully understood, but genetic factors, radiation exposure, and certain inherited conditions may increase the risk of developing these tumors. Treatment options for pituitary neoplasms include surgical removal, radiation therapy, and medical management with drugs that can help control hormonal imbalances.

Thyroid hormones are hormones produced and released by the thyroid gland, a small endocrine gland located in the neck that helps regulate metabolism, growth, and development in the human body. The two main thyroid hormones are triiodothyronine (T3) and thyroxine (T4), which contain iodine atoms. These hormones play a crucial role in various bodily functions, including heart rate, body temperature, digestion, and brain development. They help regulate the rate at which your body uses energy, affects how sensitive your body is to other hormones, and plays a vital role in the development and differentiation of all cells of the human body. Thyroid hormone levels are regulated by the hypothalamus and pituitary gland through a feedback mechanism that helps maintain proper balance.

Beta-endorphins are naturally occurring opioid peptides that are produced in the brain and other parts of the body. They are synthesized from a larger precursor protein called proopiomelanocortin (POMC) and consist of 31 amino acids. Beta-endorphins have potent analgesic effects, which means they can reduce the perception of pain. They also play a role in regulating mood, emotions, and various physiological processes such as immune function and hormonal regulation.

Beta-endorphins bind to opioid receptors in the brain and other tissues, leading to a range of effects including pain relief, sedation, euphoria, and reduced anxiety. They are released in response to stress, physical activity, and certain physiological conditions such as pregnancy and lactation. Beta-endorphins have been studied for their potential therapeutic uses in the treatment of pain, addiction, and mood disorders. However, more research is needed to fully understand their mechanisms of action and potential side effects.

Physical restraint, in a medical context, refers to the use of physical force or equipment to limit a person's movements or access to their own body. This is typically done to prevent harm to the individual themselves or to others. It can include various devices such as wrist restraints, vest restraints, or bed rails. The use of physical restraints should be a last resort and must be in accordance with established guidelines and regulations to ensure the safety and rights of the patient are respected.

Anterior pituitary hormones are a group of six major hormones that are produced and released by the anterior portion (lobe) of the pituitary gland, a small endocrine gland located at the base of the brain. These hormones play crucial roles in regulating various bodily functions and activities. The six main anterior pituitary hormones are:

1. Growth Hormone (GH): Also known as somatotropin, GH is essential for normal growth and development in children and adolescents. It helps regulate body composition, metabolism, and bone density in adults.
2. Prolactin (PRL): A hormone that stimulates milk production in females after childbirth and is also involved in various reproductive and immune functions in both sexes.
3. Follicle-Stimulating Hormone (FSH): FSH regulates the development, growth, and maturation of follicles in the ovaries (in females) and sperm production in the testes (in males).
4. Luteinizing Hormone (LH): LH plays a key role in triggering ovulation in females and stimulating testosterone production in males.
5. Thyroid-Stimulating Hormone (TSH): TSH regulates the function of the thyroid gland, which is responsible for producing and releasing thyroid hormones that control metabolism and growth.
6. Adrenocorticotropic Hormone (ACTH): ACTH stimulates the adrenal glands to produce cortisol, a steroid hormone involved in stress response, metabolism, and immune function.

These anterior pituitary hormones are regulated by the hypothalamus, which is located above the pituitary gland. The hypothalamus releases releasing and inhibiting factors that control the synthesis and secretion of anterior pituitary hormones, creating a complex feedback system to maintain homeostasis in the body.

Metyrapone is a medication that is primarily used in the diagnosis and treatment of Cushing's syndrome, a condition characterized by excessive levels of cortisol hormone in the body. It works as an inhibitor of steroidogenesis, specifically blocking the enzyme 11-beta-hydroxylase, which is involved in the production of cortisol in the adrenal gland.

By inhibiting this enzyme, metyrapone prevents the formation of cortisol and leads to an accumulation of its precursor, 11-deoxycortisol. This can help restore the balance of hormones in the body and alleviate symptoms associated with Cushing's syndrome.

It is important to note that metyrapone should only be used under the supervision of a healthcare professional, as it can have significant side effects and interactions with other medications.

Hypophysectomy is a surgical procedure that involves the removal or partial removal of the pituitary gland, also known as the hypophysis. The pituitary gland is a small endocrine gland located at the base of the brain, just above the nasal cavity, and is responsible for producing and secreting several important hormones that regulate various bodily functions.

Hypophysectomy may be performed for therapeutic or diagnostic purposes. In some cases, it may be used to treat pituitary tumors or other conditions that affect the function of the pituitary gland. It may also be performed as a research procedure in animal models to study the effects of pituitary hormone deficiency on various physiological processes.

The surgical approach for hypophysectomy may vary depending on the specific indication and the patient's individual anatomy. In general, however, the procedure involves making an incision in the skull and exposing the pituitary gland through a small opening in the bone. The gland is then carefully dissected and removed or partially removed as necessary.

Potential complications of hypophysectomy include damage to surrounding structures such as the optic nerves, which can lead to vision loss, and cerebrospinal fluid leaks. Additionally, removal of the pituitary gland can result in hormonal imbalances that may require long-term management with hormone replacement therapy.

The anterior pituitary, also known as the adenohypophysis, is the front portion of the pituitary gland. It is responsible for producing and secreting several important hormones that regulate various bodily functions. These hormones include:

* Growth hormone (GH), which stimulates growth and cell reproduction in bones and other tissues.
* Thyroid-stimulating hormone (TSH), which regulates the production of thyroid hormones by the thyroid gland.
* Adrenocorticotropic hormone (ACTH), which stimulates the adrenal glands to produce cortisol and other steroid hormones.
* Follicle-stimulating hormone (FSH) and luteinizing hormone (LH), which regulate reproductive function in both males and females by controlling the development and release of eggs or sperm.
* Prolactin, which stimulates milk production in pregnant and nursing women.
* Melanocyte-stimulating hormone (MSH), which regulates skin pigmentation and appetite.

The anterior pituitary gland is controlled by the hypothalamus, a small region of the brain located just above it. The hypothalamus produces releasing and inhibiting hormones that regulate the secretion of hormones from the anterior pituitary. These hormones are released into a network of blood vessels called the portal system, which carries them directly to the anterior pituitary gland.

Damage or disease of the anterior pituitary can lead to hormonal imbalances and various medical conditions, such as growth disorders, thyroid dysfunction, adrenal insufficiency, reproductive problems, and diabetes insipidus.

Adrenal cortex function tests are a group of diagnostic tests that evaluate the proper functioning of the adrenal cortex, which is the outer layer of the adrenal glands. These glands are located on top of each kidney and are responsible for producing several essential hormones. The adrenal cortex produces hormones such as cortisol, aldosterone, and androgens.

There are several types of adrenal cortex function tests, including:

1. Cortisol testing: This test measures the levels of cortisol in the blood or urine to determine if the adrenal glands are producing adequate amounts of this hormone. Cortisol helps regulate metabolism, immune response, and stress response.
2. ACTH (adrenocorticotropic hormone) stimulation test: This test measures the adrenal gland's response to ACTH, a hormone produced by the pituitary gland that stimulates the adrenal glands to produce cortisol. The test involves administering synthetic ACTH and measuring cortisol levels before and after administration.
3. Aldosterone testing: This test measures the levels of aldosterone in the blood or urine to determine if the adrenal glands are producing adequate amounts of this hormone. Aldosterone helps regulate electrolyte balance and blood pressure.
4. Dexamethasone suppression test: This test involves administering dexamethasone, a synthetic corticosteroid, to suppress cortisol production. The test measures cortisol levels before and after administration to determine if the adrenal glands are overproducing cortisol.
5. Androgen testing: This test measures the levels of androgens, such as testosterone and dehydroepiandrosterone (DHEA), in the blood or urine to determine if the adrenal glands are producing excessive amounts of these hormones.

Abnormal results from adrenal cortex function tests may indicate conditions such as Addison's disease, Cushing's syndrome, congenital adrenal hyperplasia, and pheochromocytoma.

Lethargy is a state of extreme fatigue, drowsiness, and/or lack of energy. In a medical context, lethargy may refer to a reduced level of consciousness or awareness where an individual has difficulty staying awake or responding to stimuli. It can be a symptom of various medical conditions such as infections, neurological disorders, metabolic imbalances, or psychological issues. However, it is important to note that lethargy should be evaluated by a healthcare professional for proper diagnosis and treatment.

Pituitary ACTH hypersecretion, also known as Cushing's disease, is a condition characterized by the excessive production of adrenocorticotropic hormone (ACTH) from the pituitary gland. This results in an overproduction of cortisol, a steroid hormone produced by the adrenal glands, leading to a constellation of symptoms known as Cushing's syndrome.

In Cushing's disease, a benign tumor called an adenoma develops on the pituitary gland, causing it to release excess ACTH. This in turn stimulates the adrenal glands to produce more cortisol than necessary. The resulting high levels of cortisol can cause various symptoms such as weight gain, particularly around the trunk and face (central obesity), thinning of the skin, bruising, weakness, fatigue, mood changes, high blood pressure, and an increased risk of infections.

It is important to distinguish Cushing's disease from other causes of Cushing's syndrome, such as cortisol-producing adrenal tumors or exogenous sources of corticosteroid use, as the treatment approach may differ. Treatment for Cushing's disease typically involves surgical removal of the pituitary tumor, with additional medical management and/or radiation therapy in some cases.

Alpha-MSH (α-MSH) stands for alpha-melanocyte stimulating hormone. It is a peptide hormone that is produced in the pituitary gland and other tissues in the body. Alpha-MSH plays a role in various physiological processes, including:

1. Melanin production: Alpha-MSH stimulates melanin production in the skin, which leads to skin tanning.
2. Appetite regulation: Alpha-MSH acts as a appetite suppressant by signaling to the brain that the stomach is full.
3. Inflammation and immune response: Alpha-MSH has anti-inflammatory effects and helps regulate the immune response.
4. Energy balance and metabolism: Alpha-MSH helps regulate energy balance and metabolism by signaling to the brain to increase or decrease food intake and energy expenditure.

Alpha-MSH exerts its effects by binding to melanocortin receptors, specifically MC1R, MC3R, MC4R, and MC5R. Dysregulation of alpha-MSH signaling has been implicated in various medical conditions, including obesity, anorexia nervosa, and certain skin disorders.

Physiological stress is a response of the body to a demand or threat that disrupts homeostasis and activates the autonomic nervous system and hypothalamic-pituitary-adrenal (HPA) axis. This results in the release of stress hormones such as adrenaline, cortisol, and noradrenaline, which prepare the body for a "fight or flight" response. Increased heart rate, rapid breathing, heightened sensory perception, and increased alertness are some of the physiological changes that occur during this response. Chronic stress can have negative effects on various bodily functions, including the immune, cardiovascular, and nervous systems.

Adrenalectomy is a surgical procedure in which one or both adrenal glands are removed. The adrenal glands are small, triangular-shaped glands located on top of each kidney that produce hormones such as cortisol, aldosterone, and adrenaline (epinephrine).

There are several reasons why an adrenalectomy may be necessary. For example, the procedure may be performed to treat tumors or growths on the adrenal glands, such as pheochromocytomas, which can cause high blood pressure and other symptoms. Adrenalectomy may also be recommended for patients with Cushing's syndrome, a condition in which the body is exposed to too much cortisol, or for those with adrenal cancer.

During an adrenalectomy, the surgeon makes an incision in the abdomen or back and removes the affected gland or glands. In some cases, laparoscopic surgery may be used, which involves making several small incisions and using specialized instruments to remove the gland. After the procedure, patients may need to take hormone replacement therapy to compensate for the loss of adrenal gland function.

Gonadotropin-Releasing Hormone (GnRH), also known as Luteinizing Hormone-Releasing Hormone (LHRH), is a hormonal peptide consisting of 10 amino acids. It is produced and released by the hypothalamus, an area in the brain that links the nervous system to the endocrine system via the pituitary gland.

GnRH plays a crucial role in regulating reproduction and sexual development through its control of two gonadotropins: follicle-stimulating hormone (FSH) and luteinizing hormone (LH). These gonadotropins, in turn, stimulate the gonads (ovaries or testes) to produce sex steroids and eggs or sperm.

GnRH acts on the anterior pituitary gland by binding to its specific receptors, leading to the release of FSH and LH. The hypothalamic-pituitary-gonadal axis is under negative feedback control, meaning that when sex steroid levels are high, they inhibit the release of GnRH, which subsequently decreases FSH and LH secretion.

GnRH agonists and antagonists have clinical applications in various medical conditions, such as infertility treatments, precocious puberty, endometriosis, uterine fibroids, prostate cancer, and hormone-responsive breast cancer.

The Zona Fasciculata is a region within the adrenal gland, which is a small gland located on top of the kidneys. It plays an essential role in endocrine function. The adrenal gland is divided into two main parts: the outer cortex and the inner medulla. The cortex itself is further divided into three zones: the Zona Glomerulosa, the Zona Fasciculata, and the Zona Reticularis.

The Zona Fasciculata is the middle layer of the adrenal cortex. It is primarily responsible for producing and releasing steroid hormones, particularly glucocorticoids such as cortisol. Cortisol helps regulate metabolism, immune response, and stress response, among other functions. The Zona Fasciculata contains large, column-shaped cells called fasciculated cells that contain lipid droplets filled with cholesterol esters. These cells convert cholesterol into pregnenolone, which is then converted into cortisol through a series of enzymatic reactions.

In summary, the Zona Fasciculata is a crucial region within the adrenal gland that produces and releases cortisol, a vital glucocorticoid hormone involved in various physiological processes.

Glucocorticoids are a class of steroid hormones that are naturally produced in the adrenal gland, or can be synthetically manufactured. They play an essential role in the metabolism of carbohydrates, proteins, and fats, and have significant anti-inflammatory effects. Glucocorticoids suppress immune responses and inflammation by inhibiting the release of inflammatory mediators from various cells, such as mast cells, eosinophils, and lymphocytes. They are frequently used in medical treatment for a wide range of conditions, including allergies, asthma, rheumatoid arthritis, dermatological disorders, and certain cancers. Prolonged use or high doses of glucocorticoids can lead to several side effects, such as weight gain, mood changes, osteoporosis, and increased susceptibility to infections.

An "apudoma" is a term that refers to a type of neuroendocrine tumor that originates from cells known as "APUD (Amine Precursor Uptake and Decarboxylation) cells." These cells are capable of taking up and decarboxylating amine precursors, which are substances that can be converted into neurotransmitters or hormones.

Apudomas can occur in various organs throughout the body, including the pancreas, lung, thyroid, and gastrointestinal tract. Some examples of apudomas include:

* Pancreatic neuroendocrine tumors (PNETs) or islet cell tumors
* Small cell lung cancer
* Medullary thyroid carcinoma
* Merkel cell carcinoma
* Carcinoid tumors

These tumors can produce and secrete a variety of hormones and neurotransmitters, leading to a range of clinical symptoms. Treatment options for apudomas may include surgery, radiation therapy, chemotherapy, or targeted therapies that are designed to specifically target the abnormal cells.

Pituitary-adrenal function tests are a group of diagnostic tests that evaluate the functioning of the pituitary gland and the adrenal gland. These glands are important components of the endocrine system, which regulates various bodily functions through the production of hormones.

The pituitary gland, located at the base of the brain, produces several hormones that regulate the function of other glands in the body, including the adrenal glands. The adrenal glands, located on top of the kidneys, produce a variety of hormones that help regulate metabolism, immune system function, blood pressure, and stress responses.

Pituitary-adrenal function tests typically include:

1. Cortisol levels: Cortisol is a hormone produced by the adrenal glands in response to stress. Blood or saliva samples may be taken at different times of the day to measure cortisol levels and evaluate the body's response to stress.
2. ACTH (adrenocorticotropic hormone) levels: ACTH is a hormone produced by the pituitary gland that stimulates the adrenal glands to produce cortisol. Blood samples may be taken to measure ACTH levels and evaluate the communication between the pituitary and adrenal glands.
3. CRH (corticotropin-releasing hormone) stimulation test: This test involves administering CRH, a hormone produced by the hypothalamus that stimulates the release of ACTH, and measuring the body's response in terms of cortisol and ACTH levels.
4. Insulin tolerance test: This test involves administering insulin to lower blood sugar levels and measuring the body's response in terms of cortisol and growth hormone levels.
5. Metyrapone or dexamethasone suppression tests: These tests involve administering medications that suppress cortisol production and measuring the body's response in terms of cortisol and ACTH levels.

These tests can help diagnose various conditions related to pituitary and adrenal gland dysfunction, such as Cushing's syndrome, Addison's disease, and hypopituitarism.

11-Hydroxycorticosteroids are a class of steroid hormones that are produced in the adrenal gland. They are created when cortisol, a type of glucocorticoid hormone, is metabolized by the enzyme 11-beta-hydroxysteroid dehydrogenase type 2 (11-β-HSD2) in the kidneys. This results in the formation of cortisone, which is then converted back to cortisol as needed.

11-Hydroxycorticosteroids are important for regulating a variety of physiological processes, including metabolism, immune response, and stress response. They also have anti-inflammatory effects and are sometimes used in medical treatments to reduce inflammation and suppress the immune system.

Elevated levels of 11-hydroxycorticosteroids can indicate an overactive adrenal gland or a tumor that is producing excess cortisol. Low levels may be seen in conditions such as Addison's disease, which is characterized by underactivity of the adrenal gland.

Medical definitions of 11-hydroxycorticosteroids typically refer to the measurement of these hormones in urine or blood tests, which can help diagnose and monitor various medical conditions.

Parathyroid hormone (PTH) is a polypeptide hormone that plays a crucial role in the regulation of calcium and phosphate levels in the body. It is produced and secreted by the parathyroid glands, which are four small endocrine glands located on the back surface of the thyroid gland.

The primary function of PTH is to maintain normal calcium levels in the blood by increasing calcium absorption from the gut, mobilizing calcium from bones, and decreasing calcium excretion by the kidneys. PTH also increases phosphate excretion by the kidneys, which helps to lower serum phosphate levels.

In addition to its role in calcium and phosphate homeostasis, PTH has been shown to have anabolic effects on bone tissue, stimulating bone formation and preventing bone loss. However, chronic elevations in PTH levels can lead to excessive bone resorption and osteoporosis.

Overall, Parathyroid Hormone is a critical hormone that helps maintain mineral homeostasis and supports healthy bone metabolism.

Melanocortin receptors (MCRs) are a group of G protein-coupled receptors that bind melanocortin peptides, which include α-, β-, and γ-melanocyte stimulating hormones (MSH) and adrenocorticotropic hormone (ACTH). These receptors are involved in a variety of physiological processes, including pigmentation, energy homeostasis, sexual function, and inflammation. There are five subtypes of melanocortin receptors (MCR1-5) that are expressed in different tissues and have distinct functions.

MCR1 is primarily expressed in melanocytes and plays a crucial role in skin and hair pigmentation. Activation of MCR1 by α-MSH leads to the production and distribution of eumelanin, which results in darker skin and hair.

MCR2 is widely expressed in the central nervous system (CNS) and peripheral tissues, including the adrenal gland, testis, and ovary. It is involved in various functions such as sexual function, feeding behavior, and energy homeostasis.

MCR3 is primarily expressed in the adrenal gland and plays a critical role in the regulation of steroid hormone production and release. Activation of MCR3 by ACTH leads to the synthesis and secretion of cortisol and other steroid hormones.

MCR4 is widely expressed in the CNS, peripheral tissues, and immune cells. It is involved in various functions such as energy homeostasis, feeding behavior, sexual function, and inflammation.

MCR5 is primarily expressed in the testis and plays a role in spermatogenesis and fertility.

Overall, melanocortin receptors are important regulators of various physiological processes, and dysregulation of these receptors has been implicated in several diseases, including obesity, metabolic disorders, and skin disorders.

Human Growth Hormone (HGH), also known as somatotropin, is a peptide hormone produced in the pituitary gland. It plays a crucial role in human development and growth by stimulating the production of another hormone called insulin-like growth factor 1 (IGF-1). IGF-1 promotes the growth and reproduction of cells throughout the body, particularly in bones and other tissues. HGH also helps regulate body composition, body fluids, muscle and bone growth, sugar and fat metabolism, and possibly heart function. It is essential for human development and continues to have important effects throughout life. The secretion of HGH decreases with age, which is thought to contribute to the aging process.

Prolactin is a hormone produced by the pituitary gland, a small gland located at the base of the brain. Its primary function is to stimulate milk production in women after childbirth, a process known as lactation. However, prolactin also plays other roles in the body, including regulating immune responses, metabolism, and behavior. In men, prolactin helps maintain the sexual glands and contributes to paternal behaviors.

Prolactin levels are usually low in both men and non-pregnant women but increase significantly during pregnancy and after childbirth. Various factors can affect prolactin levels, including stress, sleep, exercise, and certain medications. High prolactin levels can lead to medical conditions such as amenorrhea (absence of menstruation), galactorrhea (spontaneous milk production not related to childbirth), infertility, and reduced sexual desire in both men and women.

17-α-Hydroxyprogesterone is a naturally occurring hormone produced by the adrenal glands and, in smaller amounts, by the ovaries and testes. It is an intermediate in the biosynthesis of steroid hormones, including cortisol, aldosterone, and sex hormones such as testosterone and estrogen.

In a medical context, 17-α-Hydroxyprogesterone may also refer to a synthetic form of this hormone that is used in the treatment of certain medical conditions. For example, a medication called 17-alpha-hydroxyprogesterone caproate (17-OHP) is used to reduce the risk of preterm birth in women who have previously given birth prematurely. It works by suppressing uterine contractions and promoting fetal lung maturity.

It's important to note that 17-alpha-Hydroxyprogesterone should only be used under the supervision of a healthcare provider, as it can have side effects and may interact with other medications.

Hypopituitarism is a medical condition characterized by deficient secretion of one or more hormones produced by the pituitary gland, a small endocrine gland located at the base of the brain. The pituitary gland controls several other endocrine glands in the body, including the thyroid, adrenals, and sex glands (ovaries and testes).

Hypopituitarism can result from damage to the pituitary gland due to various causes such as tumors, surgery, radiation therapy, trauma, or inflammation. In some cases, hypopituitarism may also be caused by a dysfunction of the hypothalamus, a region in the brain that regulates the pituitary gland's function.

The symptoms and signs of hypopituitarism depend on which hormones are deficient and can include fatigue, weakness, decreased appetite, weight loss, low blood pressure, decreased sex drive, infertility, irregular menstrual periods, intolerance to cold, constipation, thinning hair, dry skin, and depression.

Treatment of hypopituitarism typically involves hormone replacement therapy to restore the deficient hormones' normal levels. The type and dosage of hormones used will depend on which hormones are deficient and may require regular monitoring and adjustments over time.

Pituitary function tests are a group of diagnostic exams that evaluate the proper functioning of the pituitary gland, a small endocrine gland located at the base of the brain. The pituitary gland is responsible for producing and releasing several essential hormones that regulate various bodily functions, including growth, metabolism, stress response, reproduction, and lactation.

These tests typically involve measuring the levels of different hormones in the blood, stimulating or suppressing the pituitary gland with specific medications, and assessing the body's response to these challenges. Some common pituitary function tests include:

1. Growth hormone (GH) testing: Measures GH levels in the blood, often after a provocative test using substances like insulin, arginine, clonidine, or glucagon to stimulate GH release.
2. Thyroid-stimulating hormone (TSH) and free thyroxine (FT4) testing: Assesses the function of the thyroid gland by measuring TSH and FT4 levels in response to TRH (thyrotropin-releasing hormone) stimulation.
3. Adrenocorticotropic hormone (ACTH) and cortisol testing: Evaluates the hypothalamic-pituitary-adrenal axis by measuring ACTH and cortisol levels after a CRH (corticotropin-releasing hormone) stimulation test or an insulin tolerance test.
4. Prolactin (PRL) testing: Measures PRL levels in the blood, which can be elevated due to pituitary tumors or other conditions affecting the hypothalamus.
5. Follicle-stimulating hormone (FSH) and luteinizing hormone (LH) testing: Assesses reproductive function by measuring FSH and LH levels, often in conjunction with estradiol or testosterone levels.
6. Gonadotropin-releasing hormone (GnRH) stimulation test: Evaluates gonadal function by measuring FSH and LH levels after GnRH administration.
7. Growth hormone (GH) testing: Measures GH levels in response to various stimuli, such as insulin-like growth factor-1 (IGF-1), glucagon, or arginine.
8. Vasopressin (ADH) testing: Assesses the posterior pituitary function by measuring ADH levels and performing a water deprivation test.

These tests can help diagnose various pituitary disorders, such as hypopituitarism, hyperpituitarism, or pituitary tumors, and guide appropriate treatment strategies.

Gonadal steroid hormones, also known as gonadal sex steroids, are hormones that are produced and released by the gonads (i.e., ovaries in women and testes in men). These hormones play a critical role in the development and maintenance of secondary sexual characteristics, reproductive function, and overall health.

The three main classes of gonadal steroid hormones are:

1. Androgens: These are male sex hormones that are primarily produced by the testes but also produced in smaller amounts by the ovaries and adrenal glands. The most well-known androgen is testosterone, which plays a key role in the development of male secondary sexual characteristics such as facial hair, deepening of the voice, and increased muscle mass.
2. Estrogens: These are female sex hormones that are primarily produced by the ovaries but also produced in smaller amounts by the adrenal glands. The most well-known estrogen is estradiol, which plays a key role in the development of female secondary sexual characteristics such as breast development and the menstrual cycle.
3. Progestogens: These are hormones that are produced by the ovaries during the second half of the menstrual cycle and play a key role in preparing the uterus for pregnancy. The most well-known progestogen is progesterone, which also plays a role in maintaining pregnancy and regulating the menstrual cycle.

Gonadal steroid hormones can have significant effects on various physiological processes, including bone density, cognitive function, mood, and sexual behavior. Disorders of gonadal steroid hormone production or action can lead to a range of health problems, including infertility, osteoporosis, and sexual dysfunction.

Radioimmunoassay (RIA) is a highly sensitive analytical technique used in clinical and research laboratories to measure concentrations of various substances, such as hormones, vitamins, drugs, or tumor markers, in biological samples like blood, urine, or tissues. The method relies on the specific interaction between an antibody and its corresponding antigen, combined with the use of radioisotopes to quantify the amount of bound antigen.

In a typical RIA procedure, a known quantity of a radiolabeled antigen (also called tracer) is added to a sample containing an unknown concentration of the same unlabeled antigen. The mixture is then incubated with a specific antibody that binds to the antigen. During the incubation period, the antibody forms complexes with both the radiolabeled and unlabeled antigens.

After the incubation, the unbound (free) radiolabeled antigen is separated from the antibody-antigen complexes, usually through a precipitation or separation step involving centrifugation, filtration, or chromatography. The amount of radioactivity in the pellet (containing the antibody-antigen complexes) is then measured using a gamma counter or other suitable radiation detection device.

The concentration of the unlabeled antigen in the sample can be determined by comparing the ratio of bound to free radiolabeled antigen in the sample to a standard curve generated from known concentrations of unlabeled antigen and their corresponding bound/free ratios. The higher the concentration of unlabeled antigen in the sample, the lower the amount of radiolabeled antigen that will bind to the antibody, resulting in a lower bound/free ratio.

Radioimmunoassays offer high sensitivity, specificity, and accuracy, making them valuable tools for detecting and quantifying low levels of various substances in biological samples. However, due to concerns about radiation safety and waste disposal, alternative non-isotopic immunoassay techniques like enzyme-linked immunosorbent assays (ELISAs) have become more popular in recent years.

Aldosterone is a hormone produced by the adrenal gland. It plays a key role in regulating sodium and potassium balance and maintaining blood pressure through its effects on the kidneys. Aldosterone promotes the reabsorption of sodium ions and the excretion of potassium ions in the distal tubules and collecting ducts of the nephrons in the kidneys. This increases the osmotic pressure in the blood, which in turn leads to water retention and an increase in blood volume and blood pressure.

Aldosterone is released from the adrenal gland in response to a variety of stimuli, including angiotensin II (a peptide hormone produced as part of the renin-angiotensin-aldosterone system), potassium ions, and adrenocorticotropic hormone (ACTH) from the pituitary gland. The production of aldosterone is regulated by a negative feedback mechanism involving sodium levels in the blood. High sodium levels inhibit the release of aldosterone, while low sodium levels stimulate its release.

In addition to its role in maintaining fluid and electrolyte balance and blood pressure, aldosterone has been implicated in various pathological conditions, including hypertension, heart failure, and primary hyperaldosteronism (a condition characterized by excessive production of aldosterone).

Dexamethasone is a type of corticosteroid medication, which is a synthetic version of a natural hormone produced by the adrenal glands. It is often used to reduce inflammation and suppress the immune system in a variety of medical conditions, including allergies, asthma, rheumatoid arthritis, and certain skin conditions.

Dexamethasone works by binding to specific receptors in cells, which triggers a range of anti-inflammatory effects. These include reducing the production of chemicals that cause inflammation, suppressing the activity of immune cells, and stabilizing cell membranes.

In addition to its anti-inflammatory effects, dexamethasone can also be used to treat other medical conditions, such as certain types of cancer, brain swelling, and adrenal insufficiency. It is available in a variety of forms, including tablets, liquids, creams, and injectable solutions.

Like all medications, dexamethasone can have side effects, particularly if used for long periods of time or at high doses. These may include mood changes, increased appetite, weight gain, acne, thinning skin, easy bruising, and an increased risk of infections. It is important to follow the instructions of a healthcare provider when taking dexamethasone to minimize the risk of side effects.

Cortisone is a type of corticosteroid hormone that is produced naturally in the body by the adrenal gland. It is released in response to stress and helps to regulate metabolism, reduce inflammation, and suppress the immune system. Cortisone can also be synthetically produced and is often used as a medication to treat a variety of conditions such as arthritis, asthma, and skin disorders. It works by mimicking the effects of the natural hormone in the body and reducing inflammation and suppressing the immune system. Cortisone can be administered through various routes, including oral, injectable, topical, and inhalational.

The pituitary gland is divided into three lobes: the anterior lobe (adenohypophysis), the posterior lobe (neurohypophysis), and the intermediate lobe (intermedia). The medical definition of 'Pituitary Gland, Intermediate' refers to this small and less defined region located between the anterior and posterior pituitary lobes.

The intermediate lobe is primarily responsible for producing and secreting several important hormones, most notably pro-opiomelanocortin (POMC)-derived peptides such as melanocyte-stimulating hormone (MSH) and endorphins. These hormones play crucial roles in various physiological processes, including skin pigmentation, energy balance, and pain modulation.

However, it is important to note that the intermediate lobe's activity and hormonal secretion are minimal in humans compared to other mammals. In fact, some researchers question whether the human intermediate lobe even functions at all under normal conditions due to its rudimentary nature. Nevertheless, understanding the structure and function of the pituitary gland's intermediate lobe is essential for comparative endocrinology and may provide insights into the evolution of the pituitary gland across different species.

Corticotrophs are a type of endocrine cell found in the anterior pituitary gland. They are responsible for producing and secreting adrenocorticotropic hormone (ACTH), which regulates the function of the adrenal gland. ACTH stimulates the production and release of cortisol, a steroid hormone that helps regulate metabolism, immune response, and stress response among other functions. Corticotrophs are controlled by the hypothalamus through the release of corticotropin-releasing hormone (CRH) and vasopressin. Dysfunction of corticotrophs can lead to various endocrine disorders, such as Cushing's disease, which is characterized by excessive production of ACTH and cortisol.

The adrenal cortex hormones are a group of steroid hormones produced and released by the outer portion (cortex) of the adrenal glands, which are located on top of each kidney. These hormones play crucial roles in regulating various physiological processes, including:

1. Glucose metabolism: Cortisol helps control blood sugar levels by increasing glucose production in the liver and reducing its uptake in peripheral tissues.
2. Protein and fat metabolism: Cortisol promotes protein breakdown and fatty acid mobilization, providing essential building blocks for energy production during stressful situations.
3. Immune response regulation: Cortisol suppresses immune function to prevent overactivation and potential damage to the body during stress.
4. Cardiovascular function: Aldosterone regulates electrolyte balance and blood pressure by promoting sodium reabsorption and potassium excretion in the kidneys.
5. Sex hormone production: The adrenal cortex produces small amounts of sex hormones, such as androgens and estrogens, which contribute to sexual development and function.
6. Growth and development: Cortisol plays a role in normal growth and development by influencing the activity of growth-promoting hormones like insulin-like growth factor 1 (IGF-1).

The main adrenal cortex hormones include:

1. Glucocorticoids: Cortisol is the primary glucocorticoid, responsible for regulating metabolism and stress response.
2. Mineralocorticoids: Aldosterone is the primary mineralocorticoid, involved in electrolyte balance and blood pressure regulation.
3. Androgens: Dehydroepiandrosterone (DHEA) and its sulfate derivative (DHEAS) are the most abundant adrenal androgens, contributing to sexual development and function.
4. Estrogens: Small amounts of estrogens are produced by the adrenal cortex, mainly in women.

Disorders related to impaired adrenal cortex hormone production or regulation can lead to various clinical manifestations, such as Addison's disease (adrenal insufficiency), Cushing's syndrome (hypercortisolism), and congenital adrenal hyperplasia (CAH).

Thyrotropin, also known as thyroid-stimulating hormone (TSH), is a hormone secreted by the anterior pituitary gland. Its primary function is to regulate the production and release of thyroxine (T4) and triiodothyronine (T3) hormones from the thyroid gland. Thyrotropin binds to receptors on the surface of thyroid follicular cells, stimulating the uptake of iodide and the synthesis and release of T4 and T3. The secretion of thyrotropin is controlled by the hypothalamic-pituitary-thyroid axis: thyrotropin-releasing hormone (TRH) from the hypothalamus stimulates the release of thyrotropin, while T3 and T4 inhibit its release through a negative feedback mechanism.

Petrosal sinus sampling is a medical procedure used to help diagnose the source of hormonal hypersecretion, particularly in cases of Cushing's syndrome that are difficult to locate. The petrosal sinuses are small veins located near the pituitary gland in the brain.

During the procedure, a catheter is inserted through the patient's femoral vein and guided up to the petrosal sinuses. Samples of blood are then taken from each sinus and tested for levels of adrenocorticotropic hormone (ACTH). If there is a significant difference in ACTH levels between the two samples, it suggests that the source of the hypersecretion is likely located in the pituitary gland.

If the ACTH levels are similar in both petrosal sinuses, it may indicate an ectopic source of ACTH production outside of the pituitary gland, such as in a lung tumor. The procedure can help guide treatment decisions and determine whether surgery or other therapies are appropriate.

The hypothalamus is a small, vital region of the brain that lies just below the thalamus and forms part of the limbic system. It plays a crucial role in many important functions including:

1. Regulation of body temperature, hunger, thirst, fatigue, sleep, and circadian rhythms.
2. Production and regulation of hormones through its connection with the pituitary gland (the hypophysis). It controls the release of various hormones by producing releasing and inhibiting factors that regulate the anterior pituitary's function.
3. Emotional responses, behavior, and memory formation through its connections with the limbic system structures like the amygdala and hippocampus.
4. Autonomic nervous system regulation, which controls involuntary physiological functions such as heart rate, blood pressure, and digestion.
5. Regulation of the immune system by interacting with the autonomic nervous system.

Damage to the hypothalamus can lead to various disorders like diabetes insipidus, growth hormone deficiency, altered temperature regulation, sleep disturbances, and emotional or behavioral changes.

The Paraventricular Hypothalamic Nucleus (PVN) is a nucleus in the hypothalamus, which is a part of the brain that regulates various autonomic functions and homeostatic processes. The PVN plays a crucial role in the regulation of neuroendocrine and autonomic responses to stress, as well as the control of fluid and electrolyte balance, cardiovascular function, and energy balance.

The PVN is composed of several subdivisions, including the magnocellular and parvocellular divisions. The magnocellular neurons produce and release two neuropeptides, oxytocin and vasopressin (also known as antidiuretic hormone), into the circulation via the posterior pituitary gland. These neuropeptides play important roles in social behavior, reproduction, and fluid balance.

The parvocellular neurons, on the other hand, project to various brain regions and the pituitary gland, where they release neurotransmitters and neuropeptides that regulate the hypothalamic-pituitary-adrenal (HPA) axis, which is responsible for the stress response. The PVN also contains neurons that produce corticotropin-releasing hormone (CRH), a key neurotransmitter involved in the regulation of the HPA axis and the stress response.

Overall, the Paraventricular Hypothalamic Nucleus is an essential component of the brain's regulatory systems that help maintain homeostasis and respond to stressors. Dysfunction of the PVN has been implicated in various pathological conditions, including hypertension, obesity, and mood disorders.

Thyroid hormone receptors (THRs) are nuclear receptor proteins that bind to thyroid hormones, triiodothyronine (T3) and thyroxine (T4), and regulate gene transcription in target cells. These receptors play a crucial role in the development, growth, and metabolism of an organism by mediating the actions of thyroid hormones. THRs are encoded by genes THRA and THRB, which give rise to two major isoforms: TRα1 and TRβ1. Additionally, alternative splicing results in other isoforms with distinct tissue distributions and functions. THRs function as heterodimers with retinoid X receptors (RXRs) and bind to thyroid hormone response elements (TREs) in the regulatory regions of target genes. The binding of T3 or T4 to THRs triggers a conformational change, which leads to recruitment of coactivators or corepressors, ultimately resulting in activation or repression of gene transcription.

An ACTH-secreting pituitary adenoma is a type of tumor that develops in the pituitary gland, a small gland located at the base of the brain. This type of tumor is also known as Cushing's disease.

ACTH stands for adrenocorticotropic hormone, which is a hormone produced and released by the pituitary gland. ACTH stimulates the adrenal glands (small glands located on top of the kidneys) to produce cortisol, a steroid hormone that helps regulate metabolism, helps the body respond to stress, and suppresses inflammation.

In an ACTH-secreting pituitary adenoma, the tumor cells produce and release excessive amounts of ACTH, leading to overproduction of cortisol by the adrenal glands. This can result in a constellation of symptoms known as Cushing's syndrome, which may include weight gain (especially around the trunk), fatigue, muscle weakness, mood changes, thinning of the skin, easy bruising, and increased susceptibility to infections.

Treatment for an ACTH-secreting pituitary adenoma typically involves surgical removal of the tumor, followed by medications to manage cortisol levels if necessary. Radiation therapy may also be used in some cases.

Adrenal gland diseases refer to a group of medical conditions that affect the function or structure of the adrenal glands. The adrenal glands are small, triangular-shaped glands located on top of each kidney. They are responsible for producing several essential hormones, including cortisol, aldosterone, and adrenaline (epinephrine).

There are various types of adrenal gland diseases, some of which include:

1. Adrenal Insufficiency: A condition where the adrenal glands do not produce enough hormones, particularly cortisol and aldosterone. This can lead to symptoms such as fatigue, weight loss, low blood pressure, and skin hyperpigmentation.
2. Cushing's Syndrome: A condition characterized by an excess of cortisol in the body. It can be caused by a tumor in the pituitary gland or adrenal glands, or it can result from long-term use of steroid medications.
3. Adrenal Cancer: A rare type of cancer that affects the adrenal glands. Symptoms may include abdominal pain, weight loss, and high blood pressure.
4. Pheochromocytoma: A tumor that develops in the adrenal glands and causes an overproduction of adrenaline (epinephrine) and noradrenaline (norepinephrine). Symptoms may include high blood pressure, headaches, sweating, and anxiety.
5. Adrenal Hemorrhage: A condition where bleeding occurs in the adrenal glands, often as a result of severe trauma or infection. This can lead to adrenal insufficiency and other complications.
6. Congenital Adrenal Hyperplasia: An inherited disorder that affects the production of cortisol and other hormones in the adrenal glands. Symptoms may include ambiguous genitalia, precocious puberty, and short stature.

Treatment for adrenal gland diseases varies depending on the specific condition and its severity. Treatment options may include medication, surgery, or radiation therapy.

Neurosecretory systems are specialized components of the nervous system that produce and release chemical messengers called neurohormones. These neurohormones are released into the bloodstream and can have endocrine effects on various target organs in the body. The cells that make up neurosecretory systems, known as neurosecretory cells, are found in specific regions of the brain, such as the hypothalamus, and in peripheral nerves.

Neurosecretory systems play a critical role in regulating many physiological processes, including fluid and electrolyte balance, stress responses, growth and development, reproductive functions, and behavior. The neurohormones released by these systems can act synergistically or antagonistically to maintain homeostasis and coordinate the body's response to internal and external stimuli.

Neurosecretory cells are characterized by their ability to synthesize and store neurohormones in secretory granules, which are released upon stimulation. The release of neurohormones can be triggered by a variety of signals, including neural impulses, hormonal changes, and other physiological cues. Once released into the bloodstream, neurohormones can travel to distant target organs, where they bind to specific receptors and elicit a range of responses.

Overall, neurosecretory systems are an essential component of the neuroendocrine system, which plays a critical role in regulating many aspects of human physiology and behavior.

Dehydroepiandrosterone (DHEA) is a steroid hormone produced by the adrenal glands. It serves as a precursor to other hormones, including androgens such as testosterone and estrogens such as estradiol. DHEA levels typically peak during early adulthood and then gradually decline with age.

DHEA has been studied for its potential effects on various health conditions, including aging, cognitive function, sexual dysfunction, and certain chronic diseases. However, the evidence supporting its use for these purposes is generally limited and inconclusive. As with any supplement or medication, it's important to consult with a healthcare provider before taking DHEA to ensure safety and effectiveness.

Arginine vasopressin (AVP), also known as antidiuretic hormone (ADH), is a hormone produced in the hypothalamus and stored in the posterior pituitary gland. It plays a crucial role in regulating water balance and blood pressure in the body.

AVP acts on the kidneys to promote water reabsorption, which helps maintain adequate fluid volume and osmotic balance in the body. It also constricts blood vessels, increasing peripheral vascular resistance and thereby helping to maintain blood pressure. Additionally, AVP has been shown to have effects on cognitive function, mood regulation, and pain perception.

Deficiencies or excesses of AVP can lead to a range of medical conditions, including diabetes insipidus (characterized by excessive thirst and urination), hyponatremia (low sodium levels in the blood), and syndrome of inappropriate antidiuretic hormone secretion (SIADH).

Domperidone is a medication that belongs to the class of dopamine antagonists. It works by blocking the action of dopamine, a chemical in the brain that can cause nausea and vomiting. Domperidone is primarily used to treat symptoms of gastroesophageal reflux disease (GERD) and gastric motility disorders, including bloating, fullness, and regurgitation. It works by increasing the contractions of the stomach muscles, which helps to move food and digestive juices through the stomach more quickly.

Domperidone is available in various forms, such as tablets, suspension, and injection. The medication is generally well-tolerated, but it can cause side effects such as dry mouth, diarrhea, headache, and dizziness. In rare cases, domperidone may cause more serious side effects, including irregular heart rhythms, tremors, or muscle stiffness.

It is important to note that domperidone has a risk of causing cardiac arrhythmias, particularly at higher doses and in patients with pre-existing heart conditions. Therefore, it should be used with caution and only under the supervision of a healthcare professional.

An adenoma is a benign (noncancerous) tumor that develops from glandular epithelial cells. These types of cells are responsible for producing and releasing fluids, such as hormones or digestive enzymes, into the surrounding tissues. Adenomas can occur in various organs and glands throughout the body, including the thyroid, pituitary, adrenal, and digestive systems.

Depending on their location, adenomas may cause different symptoms or remain asymptomatic. Some common examples of adenomas include:

1. Colorectal adenoma (also known as a polyp): These growths occur in the lining of the colon or rectum and can develop into colorectal cancer if left untreated. Regular screenings, such as colonoscopies, are essential for early detection and removal of these polyps.
2. Thyroid adenoma: This type of adenoma affects the thyroid gland and may result in an overproduction or underproduction of hormones, leading to conditions like hyperthyroidism (overactive thyroid) or hypothyroidism (underactive thyroid).
3. Pituitary adenoma: These growths occur in the pituitary gland, which is located at the base of the brain and controls various hormonal functions. Depending on their size and location, pituitary adenomas can cause vision problems, headaches, or hormonal imbalances that affect growth, reproduction, and metabolism.
4. Liver adenoma: These rare benign tumors develop in the liver and may not cause any symptoms unless they become large enough to press on surrounding organs or structures. In some cases, liver adenomas can rupture and cause internal bleeding.
5. Adrenal adenoma: These growths occur in the adrenal glands, which are located above the kidneys and produce hormones that regulate stress responses, metabolism, and blood pressure. Most adrenal adenomas are nonfunctioning, meaning they do not secrete excess hormones. However, functioning adrenal adenomas can lead to conditions like Cushing's syndrome or Conn's syndrome, depending on the type of hormone being overproduced.

It is essential to monitor and manage benign tumors like adenomas to prevent potential complications, such as rupture, bleeding, or hormonal imbalances. Treatment options may include surveillance with imaging studies, medication to manage hormonal issues, or surgical removal of the tumor in certain cases.

Adrenal cortex neoplasms refer to abnormal growths (tumors) in the adrenal gland's outer layer, known as the adrenal cortex. These neoplasms can be benign or malignant (cancerous). Benign tumors are called adrenal adenomas, while cancerous tumors are called adrenocortical carcinomas.

Adrenal cortex neoplasms can produce various hormones, leading to different clinical presentations. For instance, they may cause Cushing's syndrome (characterized by excessive cortisol production), Conn's syndrome (caused by aldosterone excess), or virilization (due to androgen excess). Some tumors may not produce any hormones and are discovered incidentally during imaging studies for unrelated conditions.

The diagnosis of adrenal cortex neoplasms typically involves a combination of imaging techniques, such as CT or MRI scans, and hormonal assessments to determine if the tumor is functional or non-functional. In some cases, a biopsy may be necessary to confirm the diagnosis and differentiate between benign and malignant tumors. Treatment options depend on the type, size, location, and hormonal activity of the neoplasm and may include surgical excision, radiation therapy, chemotherapy, or a combination of these approaches.

Psychological stress is the response of an individual's mind and body to challenging or demanding situations. It can be defined as a state of emotional and physical tension resulting from adversity, demand, or change. This response can involve a variety of symptoms, including emotional, cognitive, behavioral, and physiological components.

Emotional responses may include feelings of anxiety, fear, anger, sadness, or frustration. Cognitive responses might involve difficulty concentrating, racing thoughts, or negative thinking patterns. Behaviorally, psychological stress can lead to changes in appetite, sleep patterns, social interactions, and substance use. Physiologically, the body's "fight-or-flight" response is activated, leading to increased heart rate, blood pressure, muscle tension, and other symptoms.

Psychological stress can be caused by a wide range of factors, including work or school demands, financial problems, relationship issues, traumatic events, chronic illness, and major life changes. It's important to note that what causes stress in one person may not cause stress in another, as individual perceptions and coping mechanisms play a significant role.

Chronic psychological stress can have negative effects on both mental and physical health, increasing the risk of conditions such as anxiety disorders, depression, heart disease, diabetes, and autoimmune diseases. Therefore, it's essential to identify sources of stress and develop effective coping strategies to manage and reduce its impact.

Steroids, also known as corticosteroids, are a type of hormone that the adrenal gland produces in your body. They have many functions, such as controlling the balance of salt and water in your body and helping to reduce inflammation. Steroids can also be synthetically produced and used as medications to treat a variety of conditions, including allergies, asthma, skin conditions, and autoimmune disorders.

Steroid medications are available in various forms, such as oral pills, injections, creams, and inhalers. They work by mimicking the effects of natural hormones produced by your body, reducing inflammation and suppressing the immune system's response to prevent or reduce symptoms. However, long-term use of steroids can have significant side effects, including weight gain, high blood pressure, osteoporosis, and increased risk of infections.

It is important to note that anabolic steroids are a different class of drugs that are sometimes abused for their muscle-building properties. These steroids are synthetic versions of the male hormone testosterone and can have serious health consequences when taken in large doses or without medical supervision.

Addison disease, also known as primary adrenal insufficiency or hypocortisolism, is a rare endocrine disorder characterized by the dysfunction and underproduction of hormones produced by the adrenal glands, specifically cortisol and aldosterone. The adrenal glands are located on top of the kidneys and play a crucial role in regulating various bodily functions such as metabolism, blood pressure, stress response, and immune system function.

The primary cause of Addison disease is the destruction of more than 90% of the adrenal cortex, which is the outer layer of the adrenal glands responsible for hormone production. This damage can be due to an autoimmune disorder where the body's immune system mistakenly attacks and destroys the adrenal gland tissue, infections such as tuberculosis or HIV, cancer, genetic disorders, or certain medications.

The symptoms of Addison disease often develop gradually and may include fatigue, weakness, weight loss, decreased appetite, low blood pressure, darkening of the skin, and mood changes. In some cases, an acute crisis known as acute adrenal insufficiency or Addisonian crisis can occur, which is a medical emergency characterized by sudden and severe symptoms such as extreme weakness, confusion, dehydration, vomiting, diarrhea, low blood sugar, and coma.

Diagnosis of Addison disease typically involves blood tests to measure hormone levels, imaging studies such as CT scans or MRIs to assess the adrenal glands' size and structure, and stimulation tests to evaluate the adrenal glands' function. Treatment usually involves replacing the missing hormones with medications such as hydrocortisone, fludrocortisone, and sometimes mineralocorticoids. With proper treatment and management, individuals with Addison disease can lead normal and productive lives.

Adrenal gland neoplasms refer to abnormal growths or tumors in the adrenal glands. These glands are located on top of each kidney and are responsible for producing hormones that regulate various bodily functions such as metabolism, blood pressure, and stress response. Adrenal gland neoplasms can be benign (non-cancerous) or malignant (cancerous).

Benign adrenal tumors are called adenomas and are usually small and asymptomatic. However, some adenomas may produce excessive amounts of hormones, leading to symptoms such as high blood pressure, weight gain, and mood changes.

Malignant adrenal tumors are called adrenocortical carcinomas and are rare but aggressive cancers that can spread to other parts of the body. Symptoms of adrenocortical carcinoma may include abdominal pain, weight loss, and hormonal imbalances.

It is important to diagnose and treat adrenal gland neoplasms early to prevent complications and improve outcomes. Diagnostic tests may include imaging studies such as CT scans or MRIs, as well as hormone level testing and biopsy. Treatment options may include surgery, radiation therapy, chemotherapy, or a combination of these approaches.

Testosterone is a steroid hormone that belongs to androsten class of hormones. It is primarily secreted by the Leydig cells in the testes of males and, to a lesser extent, by the ovaries and adrenal glands in females. Testosterone is the main male sex hormone and anabolic steroid. It plays a key role in the development of masculine characteristics, such as body hair and muscle mass, and contributes to bone density, fat distribution, red cell production, and sex drive. In females, testosterone contributes to sexual desire and bone health. Testosterone is synthesized from cholesterol and its production is regulated by luteinizing hormone (LH) and follicle-stimulating hormone (FSH).

Growth Hormone (GH), also known as somatotropin, is a peptide hormone secreted by the somatotroph cells in the anterior pituitary gland. It plays a crucial role in regulating growth, cell reproduction, and regeneration by stimulating the production of another hormone called insulin-like growth factor 1 (IGF-1) in the liver and other tissues. GH also has important metabolic functions, such as increasing glucose levels, enhancing protein synthesis, and reducing fat storage. Its secretion is regulated by two hypothalamic hormones: growth hormone-releasing hormone (GHRH), which stimulates its release, and somatostatin (SRIF), which inhibits its release. Abnormal levels of GH can lead to various medical conditions, such as dwarfism or gigantism if there are deficiencies or excesses, respectively.

Congenital Adrenal Hyperplasia (CAH) is a group of inherited genetic disorders that affect the adrenal glands, which are triangular-shaped glands located on top of the kidneys. The adrenal glands are responsible for producing several essential hormones, including cortisol, aldosterone, and androgens.

CAH is caused by mutations in genes that code for enzymes involved in the synthesis of these hormones. The most common form of CAH is 21-hydroxylase deficiency, which affects approximately 90% to 95% of all cases. Other less common forms of CAH include 11-beta-hydroxylase deficiency and 3-beta-hydroxysteroid dehydrogenase deficiency.

The severity of the disorder can vary widely, depending on the degree of enzyme deficiency. In severe cases, the lack of cortisol production can lead to life-threatening salt wasting and electrolyte imbalances in newborns. The excess androgens produced due to the enzyme deficiency can also cause virilization, or masculinization, of female fetuses, leading to ambiguous genitalia at birth.

In milder forms of CAH, symptoms may not appear until later in childhood or even adulthood. These may include early puberty, rapid growth followed by premature fusion of the growth plates and short stature, acne, excessive hair growth, irregular menstrual periods, and infertility.

Treatment for CAH typically involves replacing the missing hormones with medications such as hydrocortisone, fludrocortisone, and/or sex hormones. Regular monitoring of hormone levels and careful management of medication doses is essential to prevent complications such as adrenal crisis, growth suppression, and osteoporosis.

In severe cases of CAH, early diagnosis and treatment can help prevent or minimize the risk of serious health problems and improve quality of life. Genetic counseling may also be recommended for affected individuals and their families to discuss the risks of passing on the disorder to future generations.

Progesterone is a steroid hormone that is primarily produced in the ovaries during the menstrual cycle and in pregnancy. It plays an essential role in preparing the uterus for implantation of a fertilized egg and maintaining the early stages of pregnancy. Progesterone works to thicken the lining of the uterus, creating a nurturing environment for the developing embryo.

During the menstrual cycle, progesterone is produced by the corpus luteum, a temporary structure formed in the ovary after an egg has been released from a follicle during ovulation. If pregnancy does not occur, the levels of progesterone will decrease, leading to the shedding of the uterine lining and menstruation.

In addition to its reproductive functions, progesterone also has various other effects on the body, such as helping to regulate the immune system, supporting bone health, and potentially influencing mood and cognition. Progesterone can be administered medically in the form of oral pills, intramuscular injections, or vaginal suppositories for various purposes, including hormone replacement therapy, contraception, and managing certain gynecological conditions.

A chromophobe adenoma is a type of benign (non-cancerous) tumor that typically arises in the pituitary gland, which is a small endocrine gland located at the base of the brain. The term "chromophobe" refers to the appearance of the cells under a microscope - they lack pigment and have a characteristic appearance with abundant clear or lightly stained cytoplasm.

Chromophobe adenomas are slow-growing tumors that can vary in size, and they may cause symptoms due to pressure on surrounding structures or by producing excess hormones. The most common hormone produced by chromophobe adenomas is prolactin, leading to symptoms such as menstrual irregularities, milk production (galactorrhea), and decreased sexual function in women, and decreased libido, erectile dysfunction, and infertility in men.

Treatment for chromophobe adenomas typically involves surgical removal of the tumor, often through a transsphenoidal approach (through the nose and sphenoid sinus). In some cases, radiation therapy or medical management with hormone-blocking drugs may also be necessary. Regular follow-up with an endocrinologist is important to monitor for any recurrence or hormonal imbalances.

Hyperaldosteronism is a medical condition characterized by the overproduction of aldosterone, a hormone produced by the adrenal glands. Aldosterone helps regulate sodium and potassium balance and blood pressure by promoting sodium retention and potassium excretion in the kidneys.

There are two types of hyperaldosteronism: primary and secondary. Primary hyperaldosteronism is caused by an overproduction of aldosterone from an abnormality within the adrenal gland, such as a tumor (Conn's syndrome) or hyperplasia. Secondary hyperaldosteronism occurs when there is an excess production of renin, a hormone produced by the kidneys, which then stimulates the adrenal glands to produce more aldosterone. This can be caused by various conditions that affect kidney function, such as renal artery stenosis or heart failure.

Symptoms of hyperaldosteronism may include high blood pressure, low potassium levels (hypokalemia), muscle weakness, and frequent urination. Diagnosis typically involves measuring aldosterone and renin levels in the blood, as well as other tests to determine the underlying cause. Treatment depends on the type and cause of hyperaldosteronism but may include medications, surgery, or lifestyle changes.

Thyroxine (T4) is a type of hormone produced and released by the thyroid gland, a small butterfly-shaped endocrine gland located in the front of your neck. It is one of two major hormones produced by the thyroid gland, with the other being triiodothyronine (T3).

Thyroxine plays a crucial role in regulating various metabolic processes in the body, including growth, development, and energy expenditure. Specifically, T4 helps to control the rate at which your body burns calories for energy, regulates protein, fat, and carbohydrate metabolism, and influences the body's sensitivity to other hormones.

T4 is produced by combining iodine and tyrosine, an amino acid found in many foods. Once produced, T4 circulates in the bloodstream and gets converted into its active form, T3, in various tissues throughout the body. Thyroxine has a longer half-life than T3, which means it remains active in the body for a more extended period.

Abnormal levels of thyroxine can lead to various medical conditions, such as hypothyroidism (underactive thyroid) or hyperthyroidism (overactive thyroid). These conditions can cause a range of symptoms, including weight gain or loss, fatigue, mood changes, and changes in heart rate and blood pressure.

Dehydroepiandrosterone sulfate (DHEA-S) is a steroid hormone that is produced by the adrenal glands. It is a modified form of dehydroepiandrosterone (DHEA), which is converted to DHEA-S in the body for storage and later conversion back to DHEA or other steroid hormones, such as testosterone and estrogen. DHEA-S is often measured in the blood as a marker of adrenal function. It is also available as a dietary supplement, although its effectiveness for any medical purpose is not well established.

I am not aware of a medical definition for "Cortodoxone." It is possible that this term is not recognized in the field of medicine as it does not appear to be a commonly used medication, treatment, or diagnostic tool. If you have any more information about where you encountered this term or its potential meaning, I would be happy to try and provide further clarification.

In the field of medicine, "time factors" refer to the duration of symptoms or time elapsed since the onset of a medical condition, which can have significant implications for diagnosis and treatment. Understanding time factors is crucial in determining the progression of a disease, evaluating the effectiveness of treatments, and making critical decisions regarding patient care.

For example, in stroke management, "time is brain," meaning that rapid intervention within a specific time frame (usually within 4.5 hours) is essential to administering tissue plasminogen activator (tPA), a clot-busting drug that can minimize brain damage and improve patient outcomes. Similarly, in trauma care, the "golden hour" concept emphasizes the importance of providing definitive care within the first 60 minutes after injury to increase survival rates and reduce morbidity.

Time factors also play a role in monitoring the progression of chronic conditions like diabetes or heart disease, where regular follow-ups and assessments help determine appropriate treatment adjustments and prevent complications. In infectious diseases, time factors are crucial for initiating antibiotic therapy and identifying potential outbreaks to control their spread.

Overall, "time factors" encompass the significance of recognizing and acting promptly in various medical scenarios to optimize patient outcomes and provide effective care.

Vasopressin, also known as antidiuretic hormone (ADH), is a hormone that helps regulate water balance in the body. It is produced by the hypothalamus and stored in the posterior pituitary gland. When the body is dehydrated or experiencing low blood pressure, vasopressin is released into the bloodstream, where it causes the kidneys to decrease the amount of urine they produce and helps to constrict blood vessels, thereby increasing blood pressure. This helps to maintain adequate fluid volume in the body and ensure that vital organs receive an adequate supply of oxygen-rich blood. In addition to its role in water balance and blood pressure regulation, vasopressin also plays a role in social behaviors such as pair bonding and trust.

Estradiol is a type of estrogen, which is a female sex hormone. It is the most potent and dominant form of estrogen in humans. Estradiol plays a crucial role in the development and maintenance of secondary sexual characteristics in women, such as breast development and regulation of the menstrual cycle. It also helps maintain bone density, protect the lining of the uterus, and is involved in cognition and mood regulation.

Estradiol is produced primarily by the ovaries, but it can also be synthesized in smaller amounts by the adrenal glands and fat cells. In men, estradiol is produced from testosterone through a process called aromatization. Abnormal levels of estradiol can contribute to various health issues, such as hormonal imbalances, infertility, osteoporosis, and certain types of cancer.

A dose-response relationship in the context of drugs refers to the changes in the effects or symptoms that occur as the dose of a drug is increased or decreased. Generally, as the dose of a drug is increased, the severity or intensity of its effects also increases. Conversely, as the dose is decreased, the effects of the drug become less severe or may disappear altogether.

The dose-response relationship is an important concept in pharmacology and toxicology because it helps to establish the safe and effective dosage range for a drug. By understanding how changes in the dose of a drug affect its therapeutic and adverse effects, healthcare providers can optimize treatment plans for their patients while minimizing the risk of harm.

The dose-response relationship is typically depicted as a curve that shows the relationship between the dose of a drug and its effect. The shape of the curve may vary depending on the drug and the specific effect being measured. Some drugs may have a steep dose-response curve, meaning that small changes in the dose can result in large differences in the effect. Other drugs may have a more gradual dose-response curve, where larger changes in the dose are needed to produce significant effects.

In addition to helping establish safe and effective dosages, the dose-response relationship is also used to evaluate the potential therapeutic benefits and risks of new drugs during clinical trials. By systematically testing different doses of a drug in controlled studies, researchers can identify the optimal dosage range for the drug and assess its safety and efficacy.

A spasm is a sudden, involuntary contraction or tightening of a muscle, group of muscles, or a hollow organ such as the ureter or bronchi. Spasms can occur as a result of various factors including muscle fatigue, injury, irritation, or abnormal nerve activity. They can cause pain and discomfort, and in some cases, interfere with normal bodily functions. For example, a spasm in the bronchi can cause difficulty breathing, while a spasm in the ureter can cause severe pain and may lead to a kidney stone blockage. The treatment for spasms depends on the underlying cause and may include medication, physical therapy, or lifestyle changes.

Endorphins are a type of neurotransmitter, which are chemicals that transmit signals in the nervous system and brain. The term "endorphin" comes from "endogenous morphine," reflecting the fact that these substances are produced naturally within the body and have effects similar to opiate drugs like morphine.

Endorphins are released in response to stress or pain, but they also occur naturally during exercise, excitement, laughter, love, and orgasm. They work by interacting with the opiate receptors in the brain to reduce the perception of pain and promote feelings of pleasure and well-being. Endorphins also play a role in regulating various physiological processes, including appetite, mood, and sleep.

In summary, endorphins are natural painkillers and mood elevators produced by the body in response to stress, pain, or enjoyable activities.

Corticotropin-releasing hormone (CRH) receptors are a type of G protein-coupled receptor found on the surface of cells in various tissues throughout the body. They play a critical role in the regulation of the hypothalamic-pituitary-adrenal (HPA) axis, which is responsible for the body's stress response.

There are two main types of CRH receptors: CRH-R1 and CRH-R2. When CRH binds to these receptors, it triggers a series of intracellular signaling events that ultimately lead to the release of adrenocorticotropic hormone (ACTH) from the pituitary gland. ACTH then stimulates the production and release of cortisol, a steroid hormone that helps regulate metabolism, immune function, and stress response.

In addition to their role in the HPA axis, CRH receptors have been implicated in a variety of other physiological processes, including anxiety, depression, addiction, and pain perception. Dysregulation of the CRH system has been associated with several psychiatric and neurological disorders, making CRH receptors an important target for drug development in these areas.

Melanocortins are a group of peptides that are derived from the post-translational processing of the proopiomelanocortin (POMC) gene. This gene is expressed in various tissues, including the pituitary gland, hypothalamus, and skin. The POMC precursor protein is cleaved into several active peptides, including adrenocorticotropic hormone (ACTH), β-melanocyte stimulating hormone (MSH), γ-MSH, and α-MSH. These melanocortins exert their effects through binding to melanocortin receptors (MCRs), which are G protein-coupled receptors.

The different melanocortins have distinct physiological roles, but they all share some common functions, such as modulating pigmentation, energy homeostasis, and immune responses. For instance, α-MSH and β-MSH bind to MCRs in the skin and increase melanin production, leading to skin tanning. Additionally, α-MSH can act on MCRs in the hypothalamus to regulate appetite and energy expenditure. ACTH, on the other hand, primarily stimulates the release of cortisol from the adrenal gland, but it can also bind to MCRs and influence pigmentation and sexual behavior.

Overall, melanocortins are crucial signaling molecules that play a significant role in various physiological processes, and dysregulation of melanocortin signaling has been implicated in several diseases, including obesity, depression, and skin disorders.

Epinephrine, also known as adrenaline, is a hormone and a neurotransmitter that is produced in the body. It is released by the adrenal glands in response to stress or excitement, and it prepares the body for the "fight or flight" response. Epinephrine works by binding to specific receptors in the body, which causes a variety of physiological effects, including increased heart rate and blood pressure, improved muscle strength and alertness, and narrowing of the blood vessels in the skin and intestines. It is also used as a medication to treat various medical conditions, such as anaphylaxis (a severe allergic reaction), cardiac arrest, and low blood pressure.

Cyclic adenosine monophosphate (cAMP) is a key secondary messenger in many biological processes, including the regulation of metabolism, gene expression, and cellular excitability. It is synthesized from adenosine triphosphate (ATP) by the enzyme adenylyl cyclase and is degraded by the enzyme phosphodiesterase.

In the body, cAMP plays a crucial role in mediating the effects of hormones and neurotransmitters on target cells. For example, when a hormone binds to its receptor on the surface of a cell, it can activate a G protein, which in turn activates adenylyl cyclase to produce cAMP. The increased levels of cAMP then activate various effector proteins, such as protein kinases, which go on to regulate various cellular processes.

Overall, the regulation of cAMP levels is critical for maintaining proper cellular function and homeostasis, and abnormalities in cAMP signaling have been implicated in a variety of diseases, including cancer, diabetes, and neurological disorders.

Beta-lipotropin (β-LPH) is a 91-amino acid polypeptide hormone that is derived from proopiomelanocortin (POMC), along with other bioactive peptides such as adrenocorticotropic hormone (ACTH), melanocyte-stimulating hormones (MSH), and β-endorphin. It is produced and released by the anterior pituitary gland in response to stress or corticotropin-releasing hormone (CRH) stimulation.

β-Lipotropin has been found to have several physiological functions, including the regulation of lipid metabolism, appetite control, and pain perception. It also exhibits opioid activity due to its ability to bind to opioid receptors in the brain, although its potency is much lower compared to other endogenous opioids like β-endorphin.

In addition to its role as a hormone, β-lipotropin has been studied for its potential therapeutic applications, particularly in the treatment of obesity and addiction. However, further research is needed to fully understand its mechanisms and clinical efficacy.

I apologize, but "sheep" is not a term that has a medical definition. It is a common animal with the scientific name Ovis aries. If you have any medical or health-related questions, I would be happy to try and help answer those for you.

17-Hydroxycorticosteroids are a class of steroid hormones that are produced in the adrenal gland. They are formed from the metabolism of cortisol, which is a hormone that helps regulate metabolism, immune response, and stress response. 17-Hydroxycorticosteroids include compounds such as cortisone and corticosterone.

These hormones have various functions in the body, including:

* Regulation of carbohydrate, fat, and protein metabolism
* Suppression of the immune system
* Modulation of the stress response
* Influence on blood pressure and electrolyte balance

Abnormal levels of 17-hydroxycorticosteroids can indicate problems with the adrenal gland or pituitary gland, which regulates adrenal function. They are often measured in urine or blood tests to help diagnose conditions such as Cushing's syndrome (overproduction of cortisol) and Addison's disease (underproduction of cortisol).

Paraneoplastic endocrine syndromes refer to a group of hormonal and related disorders that occur as remote effects of cancer. They are caused by substances (like hormones, peptides, or antibodies) produced by the tumor, which may be benign or malignant, and can affect various organs and systems in the body. These syndromes can occur before the cancer is diagnosed, making them an important consideration for early detection and treatment of the underlying malignancy.

Examples of paraneoplastic endocrine syndromes include:

1. Syndrome of Inappropriate Antidiuretic Hormone (SIADH): This occurs when a tumor, often small cell lung cancer, produces antidiuretic hormone (ADH), leading to excessive water retention and low sodium levels in the blood.
2. Cushing's Syndrome: Excessive production of adrenocorticotropic hormone (ACTH) by a tumor, often a small cell lung cancer or pancreatic neuroendocrine tumor, can lead to increased cortisol levels and symptoms such as weight gain, muscle weakness, and mood changes.
3. Ectopic Production of Parathyroid Hormone-Related Peptide (PTHrP): This occurs when a tumor, often a squamous cell carcinoma, produces PTHrP, leading to increased calcium levels in the blood and symptoms such as bone pain, kidney stones, and confusion.
4. Hypercalcemia of Malignancy: Excessive production of calcitriol (active vitamin D) by a tumor, often a lymphoma or myeloma, can lead to increased calcium levels in the blood and symptoms such as bone pain, kidney stones, and confusion.
5. Carcinoid Syndrome: This occurs when a neuroendocrine tumor, often in the gastrointestinal tract, produces serotonin and other substances, leading to symptoms such as flushing, diarrhea, and heart problems.

It is important to note that these syndromes can also be caused by non-cancerous conditions, so a thorough evaluation is necessary to make an accurate diagnosis.

Pituitary diseases refer to a group of conditions that affect the pituitary gland, a small endocrine gland located at the base of the brain. The pituitary gland is responsible for producing and secreting several important hormones that regulate various bodily functions, including growth and development, metabolism, stress response, and reproduction.

Pituitary diseases can be classified into two main categories:

1. Pituitary tumors: These are abnormal growths in or around the pituitary gland that can affect its function. Pituitary tumors can be benign (non-cancerous) or malignant (cancerous), and they can vary in size. Some pituitary tumors produce excess hormones, leading to a variety of symptoms, while others may not produce any hormones but can still cause problems by compressing nearby structures in the brain.
2. Pituitary gland dysfunction: This refers to conditions that affect the normal function of the pituitary gland without the presence of a tumor. Examples include hypopituitarism, which is a condition characterized by decreased production of one or more pituitary hormones, and Sheehan's syndrome, which occurs when the pituitary gland is damaged due to severe blood loss during childbirth.

Symptoms of pituitary diseases can vary widely depending on the specific condition and the hormones that are affected. Treatment options may include surgery, radiation therapy, medication, or a combination of these approaches.

A carcinoid tumor is a type of slow-growing neuroendocrine tumor that usually originates in the digestive tract, particularly in the small intestine. These tumors can also arise in other areas such as the lungs, appendix, and rarely in other organs. Carcinoid tumors develop from cells of the diffuse endocrine system (also known as the neuroendocrine system) that are capable of producing hormones or biologically active amines.

Carcinoid tumors can produce and release various hormones and bioactive substances, such as serotonin, histamine, bradykinins, prostaglandins, and tachykinins, which can lead to a variety of symptoms. The most common syndrome associated with carcinoid tumors is the carcinoid syndrome, characterized by flushing, diarrhea, abdominal cramping, and wheezing or difficulty breathing.

Carcinoid tumors are typically classified as functional or nonfunctional based on whether they produce and secrete hormones that cause symptoms. Functional carcinoid tumors account for approximately 30% of cases and can lead to the development of carcinoid syndrome, while nonfunctional tumors do not produce significant amounts of hormones and are often asymptomatic until they grow large enough to cause local or distant complications.

Treatment options for carcinoid tumors depend on the location, size, and extent of the tumor, as well as whether it is functional or nonfunctional. Treatment may include surgery, medications (such as somatostatin analogs, chemotherapy, or targeted therapies), and radiation therapy. Regular follow-up with imaging studies and biochemical tests is essential to monitor for recurrence and assess treatment response.

Sprague-Dawley rats are a strain of albino laboratory rats that are widely used in scientific research. They were first developed by researchers H.H. Sprague and R.C. Dawley in the early 20th century, and have since become one of the most commonly used rat strains in biomedical research due to their relatively large size, ease of handling, and consistent genetic background.

Sprague-Dawley rats are outbred, which means that they are genetically diverse and do not suffer from the same limitations as inbred strains, which can have reduced fertility and increased susceptibility to certain diseases. They are also characterized by their docile nature and low levels of aggression, making them easier to handle and study than some other rat strains.

These rats are used in a wide variety of research areas, including toxicology, pharmacology, nutrition, cancer, and behavioral studies. Because they are genetically diverse, Sprague-Dawley rats can be used to model a range of human diseases and conditions, making them an important tool in the development of new drugs and therapies.

"Cattle" is a term used in the agricultural and veterinary fields to refer to domesticated animals of the genus *Bos*, primarily *Bos taurus* (European cattle) and *Bos indicus* (Zebu). These animals are often raised for meat, milk, leather, and labor. They are also known as bovines or cows (for females), bulls (intact males), and steers/bullocks (castrated males). However, in a strict medical definition, "cattle" does not apply to humans or other animals.

Messenger RNA (mRNA) is a type of RNA (ribonucleic acid) that carries genetic information copied from DNA in the form of a series of three-base code "words," each of which specifies a particular amino acid. This information is used by the cell's machinery to construct proteins, a process known as translation. After being transcribed from DNA, mRNA travels out of the nucleus to the ribosomes in the cytoplasm where protein synthesis occurs. Once the protein has been synthesized, the mRNA may be degraded and recycled. Post-transcriptional modifications can also occur to mRNA, such as alternative splicing and addition of a 5' cap and a poly(A) tail, which can affect its stability, localization, and translation efficiency.

Esthesioneuroblastoma, also known as olfactory neuroblastoma, is a rare type of malignant tumor that develops in the upper part of the nasal cavity, near the area responsible for the sense of smell (olfaction). It arises from the olfactory nerve cells and typically affects adults between 20 to 50 years old, although it can occur at any age.

Esthesioneuroblastomas are characterized by their aggressive growth and potential to spread to other parts of the head and neck, as well as distant organs such as the lungs, bones, and bone marrow. Symptoms may include nasal congestion, nosebleeds, loss of smell, facial pain or numbness, bulging eyes, and visual disturbances.

Diagnosis is usually made through a combination of clinical examination, imaging studies (such as MRI or CT scans), and biopsy. Treatment typically involves surgical resection of the tumor, followed by radiation therapy and/or chemotherapy to reduce the risk of recurrence. Regular follow-up care is essential due to the possibility of late relapse.

Overall, prognosis varies depending on factors such as the stage of the disease at diagnosis, the patient's age, and the effectiveness of treatment. While some individuals may experience long-term survival or even cure, others may face more aggressive tumor behavior and a higher risk of recurrence.

Steroid 11-beta-hydroxylase is a crucial enzyme involved in the steroidogenesis pathway, specifically in the synthesis of cortisol and aldosterone, which are vital hormones produced by the adrenal glands. This enzyme is encoded by the CYP11B1 gene in humans.

The enzyme's primary function is to catalyze the conversion of 11-deoxycortisol to cortisol and 11-deoxycorticosterone to aldosterone through the process of hydroxylation at the 11-beta position of the steroid molecule. Cortisol is a critical glucocorticoid hormone that helps regulate metabolism, immune response, and stress response, while aldosterone is a mineralocorticoid hormone responsible for maintaining electrolyte and fluid balance in the body.

Deficiencies or mutations in the CYP11B1 gene can lead to various disorders, such as congenital adrenal hyperplasia (CAH), which may result in impaired cortisol and aldosterone production, causing hormonal imbalances and associated symptoms.

Glucocorticoid receptors (GRs) are a type of nuclear receptor proteins found inside cells that bind to glucocorticoids, a class of steroid hormones. These receptors play an essential role in the regulation of various physiological processes, including metabolism, immune response, and stress response.

When a glucocorticoid hormone such as cortisol binds to the GR, it undergoes a conformational change that allows it to translocate into the nucleus of the cell. Once inside the nucleus, the GR acts as a transcription factor, binding to specific DNA sequences called glucocorticoid response elements (GREs) located in the promoter regions of target genes. The binding of the GR to the GRE can either activate or repress gene transcription, depending on the context and the presence of co-regulatory proteins.

Glucocorticoids have diverse effects on the body, including anti-inflammatory and immunosuppressive actions. They are commonly used in clinical settings to treat a variety of conditions such as asthma, rheumatoid arthritis, and inflammatory bowel disease. However, long-term use of glucocorticoids can lead to several side effects, including osteoporosis, weight gain, and increased risk of infections, due to the widespread effects of these hormones on multiple organ systems.

Triiodothyronine (T3) is a thyroid hormone, specifically the active form of thyroid hormone, that plays a critical role in the regulation of metabolism, growth, and development in the human body. It is produced by the thyroid gland through the iodination and coupling of the amino acid tyrosine with three atoms of iodine. T3 is more potent than its precursor, thyroxine (T4), which has four iodine atoms, as T3 binds more strongly to thyroid hormone receptors and accelerates metabolic processes at the cellular level.

In circulation, about 80% of T3 is bound to plasma proteins, while the remaining 20% is unbound or free, allowing it to enter cells and exert its biological effects. The primary functions of T3 include increasing the rate of metabolic reactions, promoting protein synthesis, enhancing sensitivity to catecholamines (e.g., adrenaline), and supporting normal brain development during fetal growth and early infancy. Imbalances in T3 levels can lead to various medical conditions, such as hypothyroidism or hyperthyroidism, which may require clinical intervention and management.

A prolactinoma is a type of pituitary tumor that produces an excess amount of the hormone prolactin, leading to various symptoms. The pituitary gland, located at the base of the brain, is responsible for producing and releasing several hormones that regulate different bodily functions. Prolactin is one such hormone, primarily known for its role in stimulating milk production in women during lactation (breastfeeding).

Prolactinoma tumors can be classified into two types: microprolactinomas and macroprolactinomas. Microprolactinomas are smaller tumors, typically less than 10 millimeters in size, while macroprolactinomas are larger tumors, generally greater than 10 millimeters in size.

The overproduction of prolactin caused by these tumors can lead to several clinical manifestations, including:

1. Galactorrhea: Unusual and often spontaneous milk production or leakage from the nipples, which can occur in both men and women who do not have a recent history of pregnancy or breastfeeding.
2. Menstrual irregularities: In women, high prolactin levels can interfere with the normal functioning of other hormones, leading to menstrual irregularities such as infrequent periods (oligomenorrhea) or absent periods (amenorrhea), and sometimes infertility.
3. Sexual dysfunction: In both men and women, high prolactin levels can cause decreased libido and sexual desire. Men may also experience erectile dysfunction and reduced sperm production.
4. Bone loss: Over time, high prolactin levels can lead to decreased bone density and an increased risk of osteoporosis due to the disruption of other hormones that regulate bone health.
5. Headaches and visual disturbances: As the tumor grows, it may put pressure on surrounding structures in the brain, leading to headaches and potential vision problems such as blurred vision or decreased peripheral vision.

Diagnosis typically involves measuring prolactin levels in the blood and performing imaging tests like an MRI (magnetic resonance imaging) scan to assess the size of the tumor. Treatment usually consists of medication to lower prolactin levels, such as dopamine agonists (e.g., bromocriptine or cabergoline), which can also help shrink the tumor. In some cases, surgery may be necessary if medication is ineffective or if the tumor is large and causing severe symptoms.

Ectopic hormone production refers to the situation when a hormone is produced in an unusual location or by a type of cell that does not typically produce it. This can occur due to various reasons such as genetic mutations, cancer, or other medical conditions. The ectopic hormone production can lead to hormonal imbalances and related symptoms, as the regulation of hormones in the body becomes disrupted.

For example, in some cases of lung cancer, the tumor cells may produce adrenocorticotropic hormone (ACTH), which is typically produced by the pituitary gland. This ectopic ACTH production can result in Cushing's syndrome, a condition characterized by symptoms such as weight gain, muscle weakness, and high blood pressure.

It's important to note that ectopic hormone production is relatively rare and usually occurs in the context of specific medical conditions. If you suspect that you or someone else may have ectopic hormone production, it's important to seek medical attention from a healthcare professional who can provide appropriate evaluation and treatment.

Juvenile hormones (JHs) are a class of sesquiterpenoid compounds that play a crucial role in the regulation of insect development, reproduction, and other physiological processes. They are primarily produced by the corpora allata, a pair of endocrine glands located in the head of insects.

JHs are essential for maintaining the larval or nymphal stage of insects, preventing the expression of adult characteristics during molting. As the concentration of JH decreases in the hemolymph (insect blood), a molt to the next developmental stage occurs, and if the insect has reached its final instar, it will metamorphose into an adult.

In addition to their role in development, JHs also influence various aspects of insect reproductive physiology, such as vitellogenesis (yolk protein synthesis), oocyte maturation, and spermatogenesis. Furthermore, JHs have been implicated in regulating diapause (a period of suspended development during unfavorable environmental conditions) and caste determination in social insects like bees and ants.

Overall, juvenile hormones are vital regulators of growth, development, and reproduction in insects, making them attractive targets for the development of novel pest management strategies.

Analysis of Variance (ANOVA) is a statistical technique used to compare the means of two or more groups and determine whether there are any significant differences between them. It is a way to analyze the variance in a dataset to determine whether the variability between groups is greater than the variability within groups, which can indicate that the groups are significantly different from one another.

ANOVA is based on the concept of partitioning the total variance in a dataset into two components: variance due to differences between group means (also known as "between-group variance") and variance due to differences within each group (also known as "within-group variance"). By comparing these two sources of variance, ANOVA can help researchers determine whether any observed differences between groups are statistically significant, or whether they could have occurred by chance.

ANOVA is a widely used technique in many areas of research, including biology, psychology, engineering, and business. It is often used to compare the means of two or more experimental groups, such as a treatment group and a control group, to determine whether the treatment had a significant effect. ANOVA can also be used to compare the means of different populations or subgroups within a population, to identify any differences that may exist between them.

Pregnancy is a physiological state or condition where a fertilized egg (zygote) successfully implants and grows in the uterus of a woman, leading to the development of an embryo and finally a fetus. This process typically spans approximately 40 weeks, divided into three trimesters, and culminates in childbirth. Throughout this period, numerous hormonal and physical changes occur to support the growing offspring, including uterine enlargement, breast development, and various maternal adaptations to ensure the fetus's optimal growth and well-being.

A circadian rhythm is a roughly 24-hour biological cycle that regulates various physiological and behavioral processes in living organisms. It is driven by the body's internal clock, which is primarily located in the suprachiasmatic nucleus (SCN) of the hypothalamus in the brain.

The circadian rhythm controls many aspects of human physiology, including sleep-wake cycles, hormone secretion, body temperature, and metabolism. It helps to synchronize these processes with the external environment, particularly the day-night cycle caused by the rotation of the Earth.

Disruptions to the circadian rhythm can have negative effects on health, leading to conditions such as insomnia, sleep disorders, depression, bipolar disorder, and even increased risk of chronic diseases like cancer, diabetes, and cardiovascular disease. Factors that can disrupt the circadian rhythm include shift work, jet lag, irregular sleep schedules, and exposure to artificial light at night.

Transcortin, also known as corticosteroid-binding globulin (CBG), is a protein found in human plasma that binds and transports cortisol, corticosterone, and other steroid hormones. It plays a crucial role in the regulation of the distribution, metabolism, and elimination of these hormones. Transcortin has a higher affinity for cortisol than corticosterone, making it the primary transporter of cortisol in the bloodstream. By binding to transcortin, cortisol is prevented from rapidly entering cells and exerting its effects, thus controlling the rate at which cortisol can interact with its target tissues.

Gouty arthritis is a type of inflammatory arthritis that occurs due to the buildup of uric acid crystals in the joints. Uric acid is a waste product that is formed when the body breaks down purines, which are substances found naturally in the body and in certain foods such as organ meats, anchovies, sardines, and beer.

In people with gouty arthritis, uric acid levels in the blood become elevated, leading to the formation of sharp, needle-like crystals that can accumulate in the joints, causing pain, inflammation, and swelling. The symptoms of gouty arthritis typically occur suddenly and may include:

* Intense pain in the affected joint, often occurring at night
* Redness, warmth, and swelling in the affected area
* Stiffness and limited mobility in the affected joint

The most commonly affected joint is the big toe, but gouty arthritis can also occur in other joints such as the ankles, knees, wrists, and fingers. Over time, repeated episodes of gouty arthritis can lead to joint damage and chronic pain. Treatment typically involves medications to reduce inflammation and manage pain, as well as lifestyle changes to lower uric acid levels in the body.

"Lavandula" is the biological name for a genus of plants in the mint family, Lamiaceae. It includes around 39 species of flowering plants native to the Old World, primarily the Mediterranean region and parts of Africa, Asia, and Europe. The most common species is Lavandula angustifolia, also known as English lavender or true lavender. These plants are well-known for their fragrant purple flowers and have been used in various applications, such as perfumes, essential oils, and herbal remedies, due to their pleasant scent and potential medicinal properties. However, it is important to note that the term "Lavandula" itself does not constitute a medical definition but rather refers to a group of plants with diverse uses and benefits.

Hormone Replacement Therapy (HRT) is a medical treatment that involves the use of hormones to replace or supplement those that the body is no longer producing or no longer producing in sufficient quantities. It is most commonly used to help manage symptoms associated with menopause and conditions related to hormonal imbalances.

In women, HRT typically involves the use of estrogen and/or progesterone to alleviate hot flashes, night sweats, vaginal dryness, and mood changes that can occur during menopause. In some cases, testosterone may also be prescribed to help improve energy levels, sex drive, and overall sense of well-being.

In men, HRT is often used to treat low testosterone levels (hypogonadism) and related symptoms such as fatigue, decreased muscle mass, and reduced sex drive.

It's important to note that while HRT can be effective in managing certain symptoms, it also carries potential risks, including an increased risk of blood clots, stroke, breast cancer (in women), and cardiovascular disease. Therefore, the decision to undergo HRT should be made carefully and discussed thoroughly with a healthcare provider.

Zona glomerulosa is a region of the adrenal gland, specifically the outer portion of the adrenal cortex. It is responsible for producing mineralocorticoids, with the principal one being aldosterone. Aldosterone helps regulate electrolyte and fluid balance in the body by increasing the reabsorption of sodium ions and water in the distal nephron of the kidney while promoting the excretion of potassium ions. This process assists in maintaining blood pressure and volume within normal ranges. The zona glomerulosa's function is primarily under the control of the renin-angiotensin-aldosterone system (RAAS).

Endocrine glands are ductless glands in the human body that release hormones directly into the bloodstream, which then carry the hormones to various tissues and organs in the body. These glands play a crucial role in regulating many of the body's functions, including metabolism, growth and development, tissue function, sexual function, reproduction, sleep, and mood.

Examples of endocrine glands include the pituitary gland, thyroid gland, parathyroid glands, adrenal glands, pineal gland, pancreas, ovaries, and testes. Each of these glands produces specific hormones that have unique effects on various target tissues in the body.

The endocrine system works closely with the nervous system to regulate many bodily functions through a complex network of feedback mechanisms. Disorders of the endocrine system can result in a wide range of symptoms and health problems, including diabetes, thyroid disease, growth disorders, and sexual dysfunction.

Kampo medicine is a traditional Japanese herbal medicine that has been officially integrated into the Japanese healthcare system since the late 19th century. It is based on traditional Chinese medicine (TCM) principles and theories, but it has evolved independently in Japan over centuries to reflect local medical needs, cultural preferences, and pharmacological research.

Kampo medicine typically involves the use of complex formulas containing multiple herbs, rather than single herbs, to address various health conditions and restore balance within the body. The formulas are often adjusted based on individual patient's symptoms, constitution, and physical condition. Kampo practitioners receive extensive training in both modern Western medicine and traditional Japanese medicine, allowing them to integrate both approaches for a more holistic treatment strategy.

Kampo has been recognized by the World Health Organization (WHO) as a valuable component of traditional medicine and is increasingly being studied in clinical trials to evaluate its efficacy and safety for various health issues, including gastrointestinal disorders, menopausal symptoms, and mental health conditions.

Ovariectomy is a surgical procedure in which one or both ovaries are removed. It is also known as "ovary removal" or "oophorectomy." This procedure is often performed as a treatment for various medical conditions, including ovarian cancer, endometriosis, uterine fibroids, and pelvic pain. Ovariectomy can also be part of a larger surgical procedure called an hysterectomy, in which the uterus is also removed.

In some cases, an ovariectomy may be performed as a preventative measure for individuals at high risk of developing ovarian cancer. This is known as a prophylactic ovariectomy. After an ovariectomy, a person will no longer have menstrual periods and will be unable to become pregnant naturally. Hormone replacement therapy may be recommended in some cases to help manage symptoms associated with the loss of hormones produced by the ovaries.

Aminoglutethimide is a medication that is primarily used to treat hormone-sensitive cancers such as breast cancer and prostate cancer. It works by blocking the production of certain hormones in the body, including estrogen and cortisol. Aminoglutethimide is an inhibitor of steroid synthesis, specifically targeting the enzymes involved in the conversion of cholesterol to steroid hormones.

The medication is available in oral form and is typically taken 2-3 times a day. Common side effects include drowsiness, dizziness, dry mouth, skin rash, and changes in appetite or weight. More serious side effects may include liver damage, severe allergic reactions, and changes in heart rhythm.

It's important to note that aminoglutethimide can interact with other medications, so it's crucial to inform your healthcare provider about all the drugs you are currently taking before starting this medication. Additionally, regular monitoring of liver function and hormone levels may be necessary during treatment with aminoglutethimide.

Nelson's syndrome is a rare condition that occurs in some patients with a history of Cushing's disease who have undergone bilateral adrenalectomy (removal of both adrenal glands). Following the surgery, these patients may develop enlargement of the pituitary gland (pituitary tumor) and increased production of ACTH (adrenocorticotropic hormone) from the remaining pituitary tissue. This results in hyperpigmentation of the skin due to the melanocyte-stimulating property of ACTH, as well as other symptoms related to hormonal imbalance. It is named after the endocrinologist Don Nelson who first described this condition in 1958.

APUD cells are a type of neuroendocrine cell that originated from the neural crest and are widely distributed throughout the body. The term "APUD" is an acronym for "Amine Precursor Uptake and Decarboxylation," which describes the ability of these cells to take up and decarboxylate amino acid precursors to produce biologically active amines, such as serotonin, histamine, and catecholamines.

APUD cells are capable of synthesizing, storing, and releasing hormones or neurotransmitters in response to various stimuli. They can be found in several endocrine and neural tissues, including the thyroid gland, adrenal medulla, pituitary gland, pancreas, lungs, and gastrointestinal tract.

In the gastrointestinal tract, APUD cells are often referred to as enterochromaffin cells or Kulchitsky cells. They play a crucial role in regulating gut motility, secretion, and blood flow through the release of hormones such as serotonin, gastrin, and somatostatin.

It's worth noting that the APUD cell concept has been largely replaced by the more comprehensive neuroendocrine system concept, which encompasses a broader range of cells with neurosecretory functions.

Growth Hormone-Releasing Hormone (GHRH) is a hormone that is produced and released by the hypothalamus, a small gland located in the brain. Its primary function is to stimulate the anterior pituitary gland to release growth hormone (GH) into the bloodstream. GH plays a crucial role in growth and development, particularly during childhood and adolescence, by promoting the growth of bones and muscles.

GHRH is a 44-amino acid peptide that binds to specific receptors on the surface of pituitary cells, triggering a series of intracellular signals that ultimately lead to the release of GH. The production and release of GHRH are regulated by various factors, including sleep, stress, exercise, and nutrition.

Abnormalities in the production or function of GHRH can lead to growth disorders, such as dwarfism or gigantism, as well as other hormonal imbalances. Therefore, understanding the role of GHRH in regulating GH release is essential for diagnosing and treating these conditions.

A melanocortin receptor (MCR) is a type of G protein-coupled receptor that binds melanocortin peptides. The melanocortin-1 receptor (MC1R) is one of five known subtypes of MCRs (MC1R-MC5R).

The MC1R is primarily expressed in melanocytes, which are pigment-producing cells located in the skin, hair follicles, and eyes. This receptor plays a crucial role in determining the type of melanin that is produced in response to environmental stimuli such as UV radiation.

Activation of the MC1R by its endogenous ligands, including α-melanocyte-stimulating hormone (α-MSH) and adrenocorticotropic hormone (ACTH), leads to the activation of adenylate cyclase and an increase in intracellular cAMP levels. This results in the activation of protein kinase A and the phosphorylation of key transcription factors, which ultimately promote the expression of genes involved in melanin synthesis.

Mutations in the MC1R gene have been associated with various pigmentation disorders, including red hair color, fair skin, and an increased risk of developing skin cancer. Additionally, polymorphisms in the MC1R gene have been linked to an increased risk of developing other diseases such as obesity and type 2 diabetes.

In the context of medical definitions, "transportation" typically refers to the movement of patients from one location to another. This can include the transfer of patients between healthcare facilities (such as from a hospital to a long-term care facility), between departments within a healthcare facility (such as from the emergency department to an inpatient unit), or to and from medical appointments.

Transportation may also refer to the movement of medical equipment, supplies, or specimens between locations. In this context, transportation ensures that necessary items are delivered to the right place at the right time, which is critical for providing high-quality patient care.

It's important to note that safe and timely transportation is essential for ensuring positive patient outcomes, reducing the risk of adverse events, and improving overall healthcare efficiency.

Steroid 21-hydroxylase, also known as CYP21A2, is a crucial enzyme involved in the synthesis of steroid hormones in the adrenal gland. Specifically, it catalyzes the conversion of 17-hydroxyprogesterone to 11-deoxycortisol and progesterone to deoxycorticosterone in the glucocorticoid and mineralocorticoid pathways, respectively.

Deficiency or mutations in this enzyme can lead to a group of genetic disorders called congenital adrenal hyperplasia (CAH), which is characterized by impaired cortisol production and disrupted hormonal balance. Depending on the severity of the deficiency, CAH can result in various symptoms such as ambiguous genitalia, precocious puberty, sexual infantilism, infertility, and increased risk of adrenal crisis.

A chemical stimulation in a medical context refers to the process of activating or enhancing physiological or psychological responses in the body using chemical substances. These chemicals can interact with receptors on cells to trigger specific reactions, such as neurotransmitters and hormones that transmit signals within the nervous system and endocrine system.

Examples of chemical stimulation include the use of medications, drugs, or supplements that affect mood, alertness, pain perception, or other bodily functions. For instance, caffeine can chemically stimulate the central nervous system to increase alertness and decrease feelings of fatigue. Similarly, certain painkillers can chemically stimulate opioid receptors in the brain to reduce the perception of pain.

It's important to note that while chemical stimulation can have therapeutic benefits, it can also have adverse effects if used improperly or in excessive amounts. Therefore, it's essential to follow proper dosing instructions and consult with a healthcare provider before using any chemical substances for stimulation purposes.

"Inbred strains of rats" are genetically identical rodents that have been produced through many generations of brother-sister mating. This results in a high degree of homozygosity, where the genes at any particular locus in the genome are identical in all members of the strain.

Inbred strains of rats are widely used in biomedical research because they provide a consistent and reproducible genetic background for studying various biological phenomena, including the effects of drugs, environmental factors, and genetic mutations on health and disease. Additionally, inbred strains can be used to create genetically modified models of human diseases by introducing specific mutations into their genomes.

Some commonly used inbred strains of rats include the Wistar Kyoto (WKY), Sprague-Dawley (SD), and Fischer 344 (F344) rat strains. Each strain has its own unique genetic characteristics, making them suitable for different types of research.

"Wistar rats" are a strain of albino rats that are widely used in laboratory research. They were developed at the Wistar Institute in Philadelphia, USA, and were first introduced in 1906. Wistar rats are outbred, which means that they are genetically diverse and do not have a fixed set of genetic characteristics like inbred strains.

Wistar rats are commonly used as animal models in biomedical research because of their size, ease of handling, and relatively low cost. They are used in a wide range of research areas, including toxicology, pharmacology, nutrition, cancer, cardiovascular disease, and behavioral studies. Wistar rats are also used in safety testing of drugs, medical devices, and other products.

Wistar rats are typically larger than many other rat strains, with males weighing between 500-700 grams and females weighing between 250-350 grams. They have a lifespan of approximately 2-3 years. Wistar rats are also known for their docile and friendly nature, making them easy to handle and work with in the laboratory setting.

"Cells, cultured" is a medical term that refers to cells that have been removed from an organism and grown in controlled laboratory conditions outside of the body. This process is called cell culture and it allows scientists to study cells in a more controlled and accessible environment than they would have inside the body. Cultured cells can be derived from a variety of sources, including tissues, organs, or fluids from humans, animals, or cell lines that have been previously established in the laboratory.

Cell culture involves several steps, including isolation of the cells from the tissue, purification and characterization of the cells, and maintenance of the cells in appropriate growth conditions. The cells are typically grown in specialized media that contain nutrients, growth factors, and other components necessary for their survival and proliferation. Cultured cells can be used for a variety of purposes, including basic research, drug development and testing, and production of biological products such as vaccines and gene therapies.

It is important to note that cultured cells may behave differently than they do in the body, and results obtained from cell culture studies may not always translate directly to human physiology or disease. Therefore, it is essential to validate findings from cell culture experiments using additional models and ultimately in clinical trials involving human subjects.

An adrenocortical adenoma is a benign tumor that arises from the cells of the adrenal cortex, which is the outer layer of the adrenal gland. These tumors can produce and release various hormones, such as cortisol, aldosterone, or androgens, depending on the type of cells they originate from.

Most adrenocortical adenomas are nonfunctioning, meaning that they do not secrete excess hormones and may not cause any symptoms. However, some functioning adenomas can produce excessive amounts of hormones, leading to a variety of clinical manifestations. For example:

* Cortisol-secreting adenomas can result in Cushing's syndrome, characterized by weight gain, muscle wasting, thin skin, easy bruising, and mood changes.
* Aldosterone-producing adenomas can cause Conn's syndrome, marked by hypertension (high blood pressure), hypokalemia (low potassium levels), and metabolic alkalosis.
* Androgen-secreting adenomas may lead to hirsutism (excessive hair growth) or virilization (development of male secondary sexual characteristics) in women.

The diagnosis of an adrenocortical adenoma typically involves imaging tests, such as CT or MRI scans, and hormonal evaluations to determine if the tumor is functioning or not. Treatment usually consists of surgical removal of the tumor, especially if it is causing hormonal imbalances or growing in size.

The Cholesterol Side-Chain Cleavage Enzyme, also known as Steroidogenic Acute Regulatory (StAR) protein or P450scc, is a complex enzymatic system that plays a crucial role in the production of steroid hormones. It is located in the inner mitochondrial membrane of steroid-producing cells, such as those found in the adrenal glands, gonads, and placenta.

The Cholesterol Side-Chain Cleavage Enzyme is responsible for converting cholesterol into pregnenolone, which is the first step in the biosynthesis of all steroid hormones, including cortisol, aldosterone, sex hormones, and vitamin D. This enzymatic complex consists of two components: a flavoprotein called NADPH-cytochrome P450 oxidoreductase, which provides electrons for the reaction, and a cytochrome P450 protein called CYP11A1, which catalyzes the actual cleavage of the cholesterol side chain.

Defects in the Cholesterol Side-Chain Cleavage Enzyme can lead to various genetic disorders, such as congenital lipoid adrenal hyperplasia (CLAH), a rare autosomal recessive disorder characterized by impaired steroidogenesis and accumulation of cholesteryl esters in the adrenal glands and gonads.

Hypothalamic hormones are a group of hormones that are produced and released by the hypothalamus, a small region at the base of the brain. These hormones play a crucial role in regulating various bodily functions, including temperature, hunger, thirst, sleep, and emotional behavior.

The hypothalamus produces two main types of hormones: releasing hormones and inhibiting hormones. Releasing hormones stimulate the pituitary gland to release its own hormones, while inhibiting hormones prevent the pituitary gland from releasing hormones.

Some examples of hypothalamic hormones include:

* Thyroid-releasing hormone (TRH), which stimulates the release of thyroid-stimulating hormone (TSH) from the pituitary gland.
* Growth hormone-releasing hormone (GHRH) and somatostatin, which regulate the release of growth hormone (GH) from the pituitary gland.
* Gonadotropin-releasing hormone (GnRH), which stimulates the release of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) from the pituitary gland, which in turn regulate reproductive function.
* Corticotropin-releasing hormone (CRH), which stimulates the release of adrenocorticotropic hormone (ACTH) from the pituitary gland, which regulates the stress response.
* Prolactin-inhibiting hormone (PIH) and prolactin-releasing hormone (PRH), which regulate the release of prolactin from the pituitary gland, which is involved in lactation.

Overall, hypothalamic hormones play a critical role in maintaining homeostasis in the body by regulating various physiological processes.

Somatostatin is a hormone that inhibits the release of several hormones and also has a role in slowing down digestion. It is produced by the body in various parts of the body, including the hypothalamus (a part of the brain), the pancreas, and the gastrointestinal tract.

Somatostatin exists in two forms: somatostatin-14 and somatostatin-28, which differ in their length. Somatostatin-14 is the predominant form found in the brain, while somatostatin-28 is the major form found in the gastrointestinal tract.

Somatostatin has a wide range of effects on various physiological processes, including:

* Inhibiting the release of several hormones such as growth hormone, insulin, glucagon, and gastrin
* Slowing down digestion by inhibiting the release of digestive enzymes from the pancreas and reducing blood flow to the gastrointestinal tract
* Regulating neurotransmission in the brain

Somatostatin is used clinically as a diagnostic tool for detecting certain types of tumors that overproduce growth hormone or other hormones, and it is also used as a treatment for some conditions such as acromegaly (a condition characterized by excessive growth hormone production) and gastrointestinal disorders.

Peptide hormones are a type of hormone consisting of short chains of amino acids known as peptides. They are produced and released by various endocrine glands and play crucial roles in regulating many physiological processes in the body, including growth and development, metabolism, stress response, and reproductive functions.

Peptide hormones exert their effects by binding to specific receptors on the surface of target cells, which triggers a series of intracellular signaling events that ultimately lead to changes in cell behavior or function. Some examples of peptide hormones include insulin, glucagon, growth hormone, prolactin, oxytocin, and vasopressin.

Peptide hormones are synthesized as larger precursor proteins called prohormones, which are cleaved by enzymes to release the active peptide hormone. They are water-soluble and cannot pass through the cell membrane, so they exert their effects through autocrine, paracrine, or endocrine mechanisms. Autocrine signaling occurs when a cell releases a hormone that binds to receptors on the same cell, while paracrine signaling involves the release of a hormone that acts on nearby cells. Endocrine signaling, on the other hand, involves the release of a hormone into the bloodstream, which then travels to distant target cells to exert its effects.

I apologize for the confusion, but "Bucladesine" is not a recognized medical term or a medication in current use in medicine. It's possible that there may be some mistake or typo in the spelling. If you have any more context about where you encountered this term, I might be able to provide a more accurate and helpful response.

Pregnenolone is defined as a steroid hormone produced in the body from cholesterol. It's often referred to as the "mother hormone" since many other hormones, including cortisol, aldosterone, progesterone, testosterone, and estrogen, are synthesized from it.

Pregnenolone is primarily produced in the adrenal glands but can also be produced in smaller amounts in the brain, skin, and sex organs (ovaries and testes). It plays a crucial role in various physiological processes such as maintaining membrane fluidity, acting as an antioxidant, and contributing to cognitive function.

However, it's important to note that while pregnenolone is a hormone, over-the-counter supplements containing this compound are not approved by the FDA for any medical use or condition. As always, consult with a healthcare provider before starting any new supplement regimen.

Organ size refers to the volume or physical measurement of an organ in the body of an individual. It can be described in terms of length, width, and height or by using specialized techniques such as imaging studies (like CT scans or MRIs) to determine the volume. The size of an organ can vary depending on factors such as age, sex, body size, and overall health status. Changes in organ size may indicate various medical conditions, including growths, inflammation, or atrophy.

I couldn't find a medical definition for "Depression, Chemical" as it is not a recognized medical term. However, I can provide you with information about chemical imbalances in the brain that are associated with depression.

Major depressive disorder (MDD), commonly referred to as depression, is believed to be caused by a combination of genetic, environmental, and physiological factors. While there is no definitive evidence that depression is solely caused by a "chemical imbalance," neurotransmitter irregularities in the brain are associated with depressive symptoms. Neurotransmitters are chemical messengers that transmit signals in the brain and other parts of the body. Some of the primary neurotransmitters involved in mood regulation include serotonin, norepinephrine, and dopamine.

In depression, it is thought that there may be alterations in the functioning of these neurotransmitter systems, leading to an imbalance. For example:

1. Serotonin: Low levels of serotonin are associated with depressive symptoms. Selective serotonin reuptake inhibitors (SSRIs), a common class of antidepressants, work by increasing the availability of serotonin in the synapse (the space between neurons) to improve communication between brain cells.
2. Norepinephrine: Imbalances in norepinephrine levels can contribute to depressive symptoms and anxiety. Norepinephrine reuptake inhibitors (NRIs), tricyclic antidepressants (TCAs), and monoamine oxidase inhibitors (MAOIs) are medications that target norepinephrine to help alleviate depression.
3. Dopamine: Deficiencies in dopamine can lead to depressive symptoms, anhedonia (the inability to feel pleasure), and motivation loss. Some antidepressants, like bupropion, work by increasing dopamine levels in the brain.

In summary, while "Chemical Depression" is not a recognized medical term, chemical imbalances in neurotransmitter systems are associated with depressive symptoms. However, depression is a complex disorder that cannot be solely attributed to a single cause or a simple chemical imbalance. It is essential to consider multiple factors when diagnosing and treating depression.

Catecholamines are a group of hormones and neurotransmitters that are derived from the amino acid tyrosine. The most well-known catecholamines are dopamine, norepinephrine (also known as noradrenaline), and epinephrine (also known as adrenaline). These hormones are produced by the adrenal glands and are released into the bloodstream in response to stress. They play important roles in the "fight or flight" response, increasing heart rate, blood pressure, and alertness. In addition to their role as hormones, catecholamines also function as neurotransmitters, transmitting signals in the nervous system. Disorders of catecholamine regulation can lead to a variety of medical conditions, including hypertension, mood disorders, and neurological disorders.

Insulin is a hormone produced by the beta cells of the pancreatic islets, primarily in response to elevated levels of glucose in the circulating blood. It plays a crucial role in regulating blood glucose levels and facilitating the uptake and utilization of glucose by peripheral tissues, such as muscle and adipose tissue, for energy production and storage. Insulin also inhibits glucose production in the liver and promotes the storage of excess glucose as glycogen or triglycerides.

Deficiency in insulin secretion or action leads to impaired glucose regulation and can result in conditions such as diabetes mellitus, characterized by chronic hyperglycemia and associated complications. Exogenous insulin is used as a replacement therapy in individuals with diabetes to help manage their blood glucose levels and prevent long-term complications.

Anti-Mullerian Hormone (AMH) is a glycoprotein hormone that belongs to the transforming growth factor-beta (TGF-β) family. It is primarily produced by the granulosa cells of developing follicles in the ovaries of females. AMH plays an essential role in female reproductive physiology, as it inhibits the recruitment and further development of primordial follicles, thereby regulating the size of the primordial follicle pool and the onset of puberty.

AMH levels are often used as a biomarker for ovarian reserve assessment in women. High AMH levels indicate a larger ovarian reserve, while low levels suggest a decreased reserve, which may be associated with reduced fertility or an earlier onset of menopause. Additionally, measuring AMH levels can help predict the response to ovarian stimulation during assisted reproductive technologies (ART) such as in vitro fertilization (IVF).

Gonadal hormones, also known as sex hormones, are steroid hormones that are primarily produced by the gonads (ovaries in females and testes in males). They play crucial roles in the development and regulation of sexual characteristics and reproductive functions. The three main types of gonadal hormones are:

1. Estrogens - predominantly produced by ovaries, they are essential for female sexual development and reproduction. The most common estrogen is estradiol, which supports the growth and maintenance of secondary sexual characteristics in women, such as breast development and wider hips. Estrogens also play a role in regulating the menstrual cycle and maintaining bone health.

2. Progesterone - primarily produced by ovaries during the menstrual cycle and pregnancy, progesterone prepares the uterus for implantation of a fertilized egg and supports the growth and development of the fetus during pregnancy. It also plays a role in regulating the menstrual cycle.

3. Androgens - produced by both ovaries and testes, but primarily by testes in males. The most common androgen is testosterone, which is essential for male sexual development and reproduction. Testosterone supports the growth and maintenance of secondary sexual characteristics in men, such as facial hair, a deeper voice, and increased muscle mass. It also plays a role in regulating sex drive (libido) and bone health in both males and females.

In summary, gonadal hormones are steroid hormones produced by the gonads that play essential roles in sexual development, reproduction, and maintaining secondary sexual characteristics.

Thyroid hormone receptors (THRs) are nuclear receptor proteins that bind to thyroid hormones and mediate their effects in target cells. There are two main types of THRs, referred to as THR alpha and THR beta. THR beta is further divided into two subtypes, THR beta1 and THR beta2.

THR beta is a type of nuclear receptor that is primarily expressed in the liver, kidney, and heart, as well as in the central nervous system. It plays an important role in regulating the metabolism of carbohydrates, lipids, and proteins, as well as in the development and function of the heart. THR beta is also involved in the regulation of body weight and energy expenditure.

THR beta1 is the predominant subtype expressed in the liver and is responsible for many of the metabolic effects of thyroid hormones in this organ. THR beta2, on the other hand, is primarily expressed in the heart and plays a role in regulating cardiac function.

Abnormalities in THR beta function can lead to various diseases, including thyroid hormone resistance, a condition in which the body's cells are unable to respond properly to thyroid hormones. This can result in symptoms such as weight gain, fatigue, and cold intolerance.

Norepinephrine, also known as noradrenaline, is a neurotransmitter and a hormone that is primarily produced in the adrenal glands and is released into the bloodstream in response to stress or physical activity. It plays a crucial role in the "fight-or-flight" response by preparing the body for action through increasing heart rate, blood pressure, respiratory rate, and glucose availability.

As a neurotransmitter, norepinephrine is involved in regulating various functions of the nervous system, including attention, perception, motivation, and arousal. It also plays a role in modulating pain perception and responding to stressful or emotional situations.

In medical settings, norepinephrine is used as a vasopressor medication to treat hypotension (low blood pressure) that can occur during septic shock, anesthesia, or other critical illnesses. It works by constricting blood vessels and increasing heart rate, which helps to improve blood pressure and perfusion of vital organs.

Glucagon is a hormone produced by the alpha cells of the pancreas. Its main function is to regulate glucose levels in the blood by stimulating the liver to convert stored glycogen into glucose, which can then be released into the bloodstream. This process helps to raise blood sugar levels when they are too low, such as during hypoglycemia.

Glucagon is a 29-amino acid polypeptide that is derived from the preproglucagon protein. It works by binding to glucagon receptors on liver cells, which triggers a series of intracellular signaling events that lead to the activation of enzymes involved in glycogen breakdown.

In addition to its role in glucose regulation, glucagon has also been shown to have other physiological effects, such as promoting lipolysis (the breakdown of fat) and inhibiting gastric acid secretion. Glucagon is often used clinically in the treatment of hypoglycemia, as well as in diagnostic tests to assess pancreatic function.

Renin is a medically recognized term and it is defined as:

"A protein (enzyme) that is produced and released by specialized cells (juxtaglomerular cells) in the kidney. Renin is a key component of the renin-angiotensin-aldosterone system (RAAS), which helps regulate blood pressure and fluid balance in the body.

When the kidney detects a decrease in blood pressure or a reduction in sodium levels, it releases renin into the bloodstream. Renin then acts on a protein called angiotensinogen, converting it to angiotensin I. Angiotensin-converting enzyme (ACE) subsequently converts angiotensin I to angiotensin II, which is a potent vasoconstrictor that narrows blood vessels and increases blood pressure.

Additionally, angiotensin II stimulates the adrenal glands to release aldosterone, a hormone that promotes sodium reabsorption in the kidneys and increases water retention, further raising blood pressure.

Therefore, renin plays a critical role in maintaining proper blood pressure and electrolyte balance in the body."

In the context of medicine and pharmacology, "kinetics" refers to the study of how a drug moves throughout the body, including its absorption, distribution, metabolism, and excretion (often abbreviated as ADME). This field is called "pharmacokinetics."

1. Absorption: This is the process of a drug moving from its site of administration into the bloodstream. Factors such as the route of administration (e.g., oral, intravenous, etc.), formulation, and individual physiological differences can affect absorption.

2. Distribution: Once a drug is in the bloodstream, it gets distributed throughout the body to various tissues and organs. This process is influenced by factors like blood flow, protein binding, and lipid solubility of the drug.

3. Metabolism: Drugs are often chemically modified in the body, typically in the liver, through processes known as metabolism. These changes can lead to the formation of active or inactive metabolites, which may then be further distributed, excreted, or undergo additional metabolic transformations.

4. Excretion: This is the process by which drugs and their metabolites are eliminated from the body, primarily through the kidneys (urine) and the liver (bile).

Understanding the kinetics of a drug is crucial for determining its optimal dosing regimen, potential interactions with other medications or foods, and any necessary adjustments for special populations like pediatric or geriatric patients, or those with impaired renal or hepatic function.

Vasopressin receptors are a type of G protein-coupled receptor that bind to and are activated by the hormone vasopressin (also known as antidiuretic hormone or ADH). There are two main types of vasopressin receptors, V1 and V2.

V1 receptors are found in various tissues throughout the body, including vascular smooth muscle, heart, liver, and kidney. Activation of V1 receptors leads to vasoconstriction (constriction of blood vessels), increased heart rate and force of heart contractions, and release of glycogen from the liver.

V2 receptors are primarily found in the kidney's collecting ducts. When activated, they increase water permeability in the collecting ducts, allowing for the reabsorption of water into the bloodstream and reducing urine production. This helps to regulate fluid balance and maintain normal blood pressure.

Abnormalities in vasopressin receptor function can contribute to various medical conditions, including hypertension, heart failure, and kidney disease.

Gestational age is the length of time that has passed since the first day of the last menstrual period (LMP) in pregnant women. It is the standard unit used to estimate the age of a pregnancy and is typically expressed in weeks. This measure is used because the exact date of conception is often not known, but the start of the last menstrual period is usually easier to recall.

It's important to note that since ovulation typically occurs around two weeks after the start of the LMP, gestational age is approximately two weeks longer than fetal age, which is the actual time elapsed since conception. Medical professionals use both gestational and fetal age to track the development and growth of the fetus during pregnancy.

Intraventricular injections are a type of medical procedure where medication is administered directly into the cerebral ventricles of the brain. The cerebral ventricles are fluid-filled spaces within the brain that contain cerebrospinal fluid (CSF). This procedure is typically used to deliver drugs that target conditions affecting the central nervous system, such as infections or tumors.

Intraventricular injections are usually performed using a thin, hollow needle that is inserted through a small hole drilled into the skull. The medication is then injected directly into the ventricles, allowing it to circulate throughout the CSF and reach the brain tissue more efficiently than other routes of administration.

This type of injection is typically reserved for situations where other methods of drug delivery are not effective or feasible. It carries a higher risk of complications, such as bleeding, infection, or damage to surrounding tissues, compared to other routes of administration. Therefore, it is usually performed by trained medical professionals in a controlled clinical setting.

"Swine" is a common term used to refer to even-toed ungulates of the family Suidae, including domestic pigs and wild boars. However, in a medical context, "swine" often appears in the phrase "swine flu," which is a strain of influenza virus that typically infects pigs but can also cause illness in humans. The 2009 H1N1 pandemic was caused by a new strain of swine-origin influenza A virus, which was commonly referred to as "swine flu." It's important to note that this virus is not transmitted through eating cooked pork products; it spreads from person to person, mainly through respiratory droplets produced when an infected person coughs or sneezes.

A fetus is the developing offspring in a mammal, from the end of the embryonic period (approximately 8 weeks after fertilization in humans) until birth. In humans, the fetal stage of development starts from the eleventh week of pregnancy and continues until childbirth, which is termed as full-term pregnancy at around 37 to 40 weeks of gestation. During this time, the organ systems become fully developed and the body grows in size. The fetus is surrounded by the amniotic fluid within the amniotic sac and is connected to the placenta via the umbilical cord, through which it receives nutrients and oxygen from the mother. Regular prenatal care is essential during this period to monitor the growth and development of the fetus and ensure a healthy pregnancy and delivery.

Androgens are a class of hormones that are primarily responsible for the development and maintenance of male sexual characteristics and reproductive function. Testosterone is the most well-known androgen, but other androgens include dehydroepiandrosterone (DHEA), androstenedione, and dihydrotestosterone (DHT).

Androgens are produced primarily by the testes in men and the ovaries in women, although small amounts are also produced by the adrenal glands in both sexes. They play a critical role in the development of male secondary sexual characteristics during puberty, such as the growth of facial hair, deepening of the voice, and increased muscle mass.

In addition to their role in sexual development and function, androgens also have important effects on bone density, mood, and cognitive function. Abnormal levels of androgens can contribute to a variety of medical conditions, including infertility, erectile dysfunction, acne, hirsutism (excessive hair growth), and prostate cancer.

I'm sorry for any confusion, but "Housing, Animal" is not a standard term in medical terminology. Medical terminology typically relates to the human body, diseases, treatments, and healthcare practices. "Housing, Animal" would be more related to veterinary medicine or animal care fields, which pertain to the accommodation and environment provided for animals. If you have any questions related to medical terminology, I'd be happy to help!

Immobilization is a medical term that refers to the restriction of normal mobility or motion of a body part, usually to promote healing and prevent further injury. This is often achieved through the use of devices such as casts, splints, braces, slings, or traction. The goal of immobilization is to keep the injured area in a fixed position so that it can heal properly without additional damage. It may be used for various medical conditions, including fractures, dislocations, sprains, strains, and soft tissue injuries. Immobilization helps reduce pain, minimize swelling, and protect the injured site from movement that could worsen the injury or impair healing.

Cycloheximide is an antibiotic that is primarily used in laboratory settings to inhibit protein synthesis in eukaryotic cells. It is derived from the actinobacteria species Streptomyces griseus. In medical terms, it is not used as a therapeutic drug in humans due to its significant side effects, including liver toxicity and potential neurotoxicity. However, it remains a valuable tool in research for studying protein function and cellular processes.

The antibiotic works by binding to the 60S subunit of the ribosome, thereby preventing the transfer RNA (tRNA) from delivering amino acids to the growing polypeptide chain during translation. This inhibition of protein synthesis can be lethal to cells, making cycloheximide a useful tool in studying cellular responses to protein depletion or misregulation.

In summary, while cycloheximide has significant research applications due to its ability to inhibit protein synthesis in eukaryotic cells, it is not used as a therapeutic drug in humans because of its toxic side effects.

Body weight is the measure of the force exerted on a scale or balance by an object's mass, most commonly expressed in units such as pounds (lb) or kilograms (kg). In the context of medical definitions, body weight typically refers to an individual's total weight, which includes their skeletal muscle, fat, organs, and bodily fluids.

Healthcare professionals often use body weight as a basic indicator of overall health status, as it can provide insights into various aspects of a person's health, such as nutritional status, metabolic function, and risk factors for certain diseases. For example, being significantly underweight or overweight can increase the risk of developing conditions like malnutrition, diabetes, heart disease, and certain types of cancer.

It is important to note that body weight alone may not provide a complete picture of an individual's health, as it does not account for factors such as muscle mass, bone density, or body composition. Therefore, healthcare professionals often use additional measures, such as body mass index (BMI), waist circumference, and blood tests, to assess overall health status more comprehensively.

Gastrointestinal (GI) hormones are a group of hormones that are secreted by cells in the gastrointestinal tract in response to food intake and digestion. They play crucial roles in regulating various physiological processes, including appetite regulation, gastric acid secretion, motility of the gastrointestinal tract, insulin secretion, and pancreatic enzyme release.

Examples of GI hormones include:

* Gastrin: Secreted by G cells in the stomach, gastrin stimulates the release of hydrochloric acid from parietal cells in the stomach lining.
* Ghrelin: Produced by the stomach, ghrelin is often referred to as the "hunger hormone" because it stimulates appetite and food intake.
* Cholecystokinin (CCK): Secreted by I cells in the small intestine, CCK promotes digestion by stimulating the release of pancreatic enzymes and bile from the liver. It also inhibits gastric emptying and reduces appetite.
* Gastric inhibitory peptide (GIP): Produced by K cells in the small intestine, GIP promotes insulin secretion and inhibits glucagon release.
* Secretin: Released by S cells in the small intestine, secretin stimulates the pancreas to produce bicarbonate-rich fluid that neutralizes stomach acid in the duodenum.
* Motilin: Secreted by MO cells in the small intestine, motilin promotes gastrointestinal motility and regulates the migrating motor complex (MMC), which is responsible for cleaning out the small intestine between meals.

These hormones work together to regulate digestion and maintain homeostasis in the body. Dysregulation of GI hormones can contribute to various gastrointestinal disorders, such as gastroparesis, irritable bowel syndrome (IBS), and diabetes.

Calcitonin is a hormone that is produced and released by the parafollicular cells (also known as C cells) of the thyroid gland. It plays a crucial role in regulating calcium homeostasis in the body. Specifically, it helps to lower elevated levels of calcium in the blood by inhibiting the activity of osteoclasts, which are bone cells that break down bone tissue and release calcium into the bloodstream. Calcitonin also promotes the uptake of calcium in the bones and increases the excretion of calcium in the urine.

Calcitonin is typically released in response to high levels of calcium in the blood, and its effects help to bring calcium levels back into balance. In addition to its role in calcium regulation, calcitonin may also have other functions in the body, such as modulating immune function and reducing inflammation.

Clinically, synthetic forms of calcitonin are sometimes used as a medication to treat conditions related to abnormal calcium levels, such as hypercalcemia (high blood calcium) or osteoporosis. Calcitonin can be administered as an injection, nasal spray, or oral tablet, depending on the specific formulation and intended use.

'Gene expression regulation' refers to the processes that control whether, when, and where a particular gene is expressed, meaning the production of a specific protein or functional RNA encoded by that gene. This complex mechanism can be influenced by various factors such as transcription factors, chromatin remodeling, DNA methylation, non-coding RNAs, and post-transcriptional modifications, among others. Proper regulation of gene expression is crucial for normal cellular function, development, and maintaining homeostasis in living organisms. Dysregulation of gene expression can lead to various diseases, including cancer and genetic disorders.

Molecular sequence data refers to the specific arrangement of molecules, most commonly nucleotides in DNA or RNA, or amino acids in proteins, that make up a biological macromolecule. This data is generated through laboratory techniques such as sequencing, and provides information about the exact order of the constituent molecules. This data is crucial in various fields of biology, including genetics, evolution, and molecular biology, allowing for comparisons between different organisms, identification of genetic variations, and studies of gene function and regulation.

Immunohistochemistry (IHC) is a technique used in pathology and laboratory medicine to identify specific proteins or antigens in tissue sections. It combines the principles of immunology and histology to detect the presence and location of these target molecules within cells and tissues. This technique utilizes antibodies that are specific to the protein or antigen of interest, which are then tagged with a detection system such as a chromogen or fluorophore. The stained tissue sections can be examined under a microscope, allowing for the visualization and analysis of the distribution and expression patterns of the target molecule in the context of the tissue architecture. Immunohistochemistry is widely used in diagnostic pathology to help identify various diseases, including cancer, infectious diseases, and immune-mediated disorders.

Reference values, also known as reference ranges or reference intervals, are the set of values that are considered normal or typical for a particular population or group of people. These values are often used in laboratory tests to help interpret test results and determine whether a patient's value falls within the expected range.

The process of establishing reference values typically involves measuring a particular biomarker or parameter in a large, healthy population and then calculating the mean and standard deviation of the measurements. Based on these statistics, a range is established that includes a certain percentage of the population (often 95%) and excludes extreme outliers.

It's important to note that reference values can vary depending on factors such as age, sex, race, and other demographic characteristics. Therefore, it's essential to use reference values that are specific to the relevant population when interpreting laboratory test results. Additionally, reference values may change over time due to advances in measurement technology or changes in the population being studied.

"Random allocation," also known as "random assignment" or "randomization," is a process used in clinical trials and other research studies to distribute participants into different intervention groups (such as experimental group vs. control group) in a way that minimizes selection bias and ensures the groups are comparable at the start of the study.

In random allocation, each participant has an equal chance of being assigned to any group, and the assignment is typically made using a computer-generated randomization schedule or other objective methods. This process helps to ensure that any differences between the groups are due to the intervention being tested rather than pre-existing differences in the participants' characteristics.

Thyrotropin-Releasing Hormone (TRH) is a tripeptide hormone that is produced and released by the hypothalamus in the brain. Its main function is to regulate the release of thyroid-stimulating hormone (TSH) from the anterior pituitary gland. TRH acts on the pituitary gland to stimulate the synthesis and secretion of TSH, which then stimulates the thyroid gland to produce and release thyroid hormones (triiodothyronine (T3) and thyroxine (T4)) into the bloodstream.

TRH is a tripeptide amino acid sequence with the structure of pGlu-His-Pro-NH2, and it is synthesized as a larger precursor molecule called preprothyrotropin-releasing hormone (preproTRH) in the hypothalamus. PreproTRH undergoes post-translational processing to produce TRH, which is then stored in secretory vesicles and released into the hypophyseal portal system, where it travels to the anterior pituitary gland and binds to TRH receptors on thyrotroph cells.

In addition to its role in regulating TSH release, TRH has been shown to have other physiological functions, including modulation of feeding behavior, body temperature, and neurotransmitter release. Dysregulation of the TRH-TSH axis can lead to various thyroid disorders, such as hypothyroidism or hyperthyroidism.

Hyperplasia is a medical term that refers to an abnormal increase in the number of cells in an organ or tissue, leading to an enlargement of the affected area. It's a response to various stimuli such as hormones, chronic irritation, or inflammation. Hyperplasia can be physiological, like the growth of breast tissue during pregnancy, or pathological, like in the case of benign or malignant tumors. The process is generally reversible if the stimulus is removed. It's important to note that hyperplasia itself is not cancerous, but some forms of hyperplasia can increase the risk of developing cancer over time.

Animal disease models are specialized animals, typically rodents such as mice or rats, that have been genetically engineered or exposed to certain conditions to develop symptoms and physiological changes similar to those seen in human diseases. These models are used in medical research to study the pathophysiology of diseases, identify potential therapeutic targets, test drug efficacy and safety, and understand disease mechanisms.

The genetic modifications can include knockout or knock-in mutations, transgenic expression of specific genes, or RNA interference techniques. The animals may also be exposed to environmental factors such as chemicals, radiation, or infectious agents to induce the disease state.

Examples of animal disease models include:

1. Mouse models of cancer: Genetically engineered mice that develop various types of tumors, allowing researchers to study cancer initiation, progression, and metastasis.
2. Alzheimer's disease models: Transgenic mice expressing mutant human genes associated with Alzheimer's disease, which exhibit amyloid plaque formation and cognitive decline.
3. Diabetes models: Obese and diabetic mouse strains like the NOD (non-obese diabetic) or db/db mice, used to study the development of type 1 and type 2 diabetes, respectively.
4. Cardiovascular disease models: Atherosclerosis-prone mice, such as ApoE-deficient or LDLR-deficient mice, that develop plaque buildup in their arteries when fed a high-fat diet.
5. Inflammatory bowel disease models: Mice with genetic mutations affecting intestinal barrier function and immune response, such as IL-10 knockout or SAMP1/YitFc mice, which develop colitis.

Animal disease models are essential tools in preclinical research, but it is important to recognize their limitations. Differences between species can affect the translatability of results from animal studies to human patients. Therefore, researchers must carefully consider the choice of model and interpret findings cautiously when applying them to human diseases.

Angiotensin II is a potent vasoactive peptide hormone that plays a critical role in the renin-angiotensin-aldosterone system (RAAS), which is a crucial regulator of blood pressure and fluid balance in the body. It is formed from angiotensin I through the action of an enzyme called angiotensin-converting enzyme (ACE).

Angiotensin II has several physiological effects on various organs, including:

1. Vasoconstriction: Angiotensin II causes contraction of vascular smooth muscle, leading to an increase in peripheral vascular resistance and blood pressure.
2. Aldosterone release: Angiotensin II stimulates the adrenal glands to release aldosterone, a hormone that promotes sodium reabsorption and potassium excretion in the kidneys, thereby increasing water retention and blood volume.
3. Sympathetic nervous system activation: Angiotensin II activates the sympathetic nervous system, leading to increased heart rate and contractility, further contributing to an increase in blood pressure.
4. Thirst regulation: Angiotensin II stimulates the hypothalamus to increase thirst, promoting water intake and helping to maintain intravascular volume.
5. Cell growth and fibrosis: Angiotensin II has been implicated in various pathological processes, such as cell growth, proliferation, and fibrosis, which can contribute to the development of cardiovascular and renal diseases.

Angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) are two classes of medications commonly used in clinical practice to target the RAAS by blocking the formation or action of angiotensin II, respectively. These drugs have been shown to be effective in managing hypertension, heart failure, and chronic kidney disease.

Thyroid hormone receptors (THRs) are nuclear receptor proteins that bind to thyroid hormones and mediate their effects in the body. There are two main types of THRs, referred to as THRα and THRβ.

THRα is a subtype of thyroid hormone receptor that is primarily expressed in tissues such as the heart, skeletal muscle, and brown adipose tissue. It plays an important role in regulating metabolism, growth, and development in these tissues. THRα has two subtypes, THRα1 and THRα2, which have different functions and are expressed in different tissues.

THRα1 is the predominant form of THRα and is found in many tissues, including the heart, skeletal muscle, and brown adipose tissue. It regulates genes involved in metabolism, growth, and development, and has been shown to play a role in regulating heart rate and contractility.

THRα2, on the other hand, is primarily expressed in the brain and pituitary gland, where it regulates the production of thyroid-stimulating hormone (TSH). THRα2 is unable to bind to thyroid hormones, but can form heterodimers with THRα1 or THRβ1, which allows it to modulate their activity.

Overall, THRα plays an important role in regulating various physiological processes in the body, and dysregulation of THRα function has been implicated in a number of diseases, including heart disease, muscle wasting, and neurological disorders.

A base sequence in the context of molecular biology refers to the specific order of nucleotides in a DNA or RNA molecule. In DNA, these nucleotides are adenine (A), guanine (G), cytosine (C), and thymine (T). In RNA, uracil (U) takes the place of thymine. The base sequence contains genetic information that is transcribed into RNA and ultimately translated into proteins. It is the exact order of these bases that determines the genetic code and thus the function of the DNA or RNA molecule.

Glycoprotein hormones are a group of hormones that share a similar structure and are made up of four subunits: two identical alpha subunits and two distinct beta subunits. The alpha subunit is common to all glycoprotein hormones, including thyroid-stimulating hormone (TSH), follicle-stimulating hormone (FSH), luteinizing hormone (LH), and human chorionic gonadotropin (hCG).

The alpha subunit of glycoprotein hormones is a 92 amino acid polypeptide chain that contains several disulfide bonds, which help to stabilize its structure. It is heavily glycosylated, meaning that it contains many carbohydrate groups attached to the protein backbone. The alpha subunit plays an important role in the biological activity of the hormone by interacting with a specific receptor on the target cell surface.

The alpha subunit contains several regions that are important for its function, including a signal peptide, a variable region, and a conserved region. The signal peptide is a short sequence of amino acids at the N-terminus of the protein that directs it to the endoplasmic reticulum for processing and secretion. The variable region contains several amino acid residues that differ between different glycoprotein hormones, while the conserved region contains amino acids that are identical or very similar in all glycoprotein hormones.

Together with the beta subunit, the alpha subunit forms the functional hormone molecule. The beta subunit determines the specificity of the hormone for its target cells and regulates its biological activity.

I believe there might be a misunderstanding in your question. "Dogs" is not a medical term or condition. It is the common name for a domesticated carnivore of the family Canidae, specifically the genus Canis, which includes wolves, foxes, and other extant and extinct species of mammals. Dogs are often kept as pets and companions, and they have been bred in a wide variety of forms and sizes for different purposes, such as hunting, herding, guarding, assisting police and military forces, and providing companionship and emotional support.

If you meant to ask about a specific medical condition or term related to dogs, please provide more context so I can give you an accurate answer.

Insect hormones are chemical messengers that regulate various physiological and behavioral processes in insects. They are produced and released by endocrine glands and organs, such as the corpora allata, prothoracic glands, and neurosecretory cells located in the brain. Insect hormones play crucial roles in the regulation of growth and development, reproduction, diapause (a state of dormancy), metamorphosis, molting, and other vital functions. Some well-known insect hormones include juvenile hormone (JH), ecdysteroids (such as 20-hydroxyecdysone), and neuropeptides like the brain hormone and adipokinetic hormone. These hormones act through specific receptors, often transmembrane proteins, to elicit intracellular signaling cascades that ultimately lead to changes in gene expression, cell behavior, or organ function. Understanding insect hormones is essential for developing novel strategies for pest management and control, as well as for advancing our knowledge of insect biology and evolution.

Blood glucose, also known as blood sugar, is the concentration of glucose in the blood. Glucose is a simple sugar that serves as the main source of energy for the body's cells. It is carried to each cell through the bloodstream and is absorbed into the cells with the help of insulin, a hormone produced by the pancreas.

The normal range for blood glucose levels in humans is typically between 70 and 130 milligrams per deciliter (mg/dL) when fasting, and less than 180 mg/dL after meals. Levels that are consistently higher than this may indicate diabetes or other metabolic disorders.

Blood glucose levels can be measured through a variety of methods, including fingerstick blood tests, continuous glucose monitoring systems, and laboratory tests. Regular monitoring of blood glucose levels is important for people with diabetes to help manage their condition and prevent complications.

"Animal pregnancy" is not a term that is typically used in medical definitions. However, in biological terms, animal pregnancy refers to the condition where a fertilized egg (or eggs) implants and develops inside the reproductive tract of a female animal, leading to the birth of offspring (live young).

The specific details of animal pregnancy can vary widely between different species, with some animals exhibiting phenomena such as placental development, gestation periods, and hormonal changes that are similar to human pregnancy, while others may have very different reproductive strategies.

It's worth noting that the study of animal pregnancy and reproduction is an important area of biological research, as it can provide insights into fundamental mechanisms of embryonic development, genetics, and evolution.

Proto-oncogene proteins, such as c-Fos, are normal cellular proteins that play crucial roles in various biological processes including cell growth, differentiation, and survival. They can be activated or overexpressed due to genetic alterations, leading to the formation of cancerous cells. The c-Fos protein is a nuclear phosphoprotein involved in signal transduction pathways and forms a heterodimer with c-Jun to create the activator protein-1 (AP-1) transcription factor complex. This complex binds to specific DNA sequences, thereby regulating the expression of target genes that contribute to various cellular responses, including proliferation, differentiation, and apoptosis. Dysregulation of c-Fos can result in uncontrolled cell growth and malignant transformation, contributing to tumor development and progression.

"Sex characteristics" refer to the anatomical, chromosomal, and genetic features that define males and females. These include both primary sex characteristics (such as reproductive organs like ovaries or testes) and secondary sex characteristics (such as breasts or facial hair) that typically develop during puberty. Sex characteristics are primarily determined by the presence of either X or Y chromosomes, with XX individuals usually developing as females and XY individuals usually developing as males, although variations and exceptions to this rule do occur.

Hormone antagonists are substances or drugs that block the action of hormones by binding to their receptors without activating them, thereby preventing the hormones from exerting their effects. They can be classified into two types: receptor antagonists and enzyme inhibitors. Receptor antagonists bind directly to hormone receptors and prevent the hormone from binding, while enzyme inhibitors block the production or breakdown of hormones by inhibiting specific enzymes involved in their metabolism. Hormone antagonists are used in the treatment of various medical conditions, such as cancer, hormonal disorders, and cardiovascular diseases.

Intravenous injections are a type of medical procedure where medication or fluids are administered directly into a vein using a needle and syringe. This route of administration is also known as an IV injection. The solution injected enters the patient's bloodstream immediately, allowing for rapid absorption and onset of action. Intravenous injections are commonly used to provide quick relief from symptoms, deliver medications that are not easily absorbed by other routes, or administer fluids and electrolytes in cases of dehydration or severe illness. It is important that intravenous injections are performed using aseptic technique to minimize the risk of infection.

... (ACTH; also adrenocorticotropin, corticotropin) is a polypeptide tropic hormone produced by and ... Adrenocorticotropic+Hormone at the U.S. National Library of Medicine Medical Subject Headings (MeSH) (Articles with short ... "Adrenocorticotropic Hormone (ACTH)". "Pro-opiomelocortin precursor". Retrieved April 8, 2013. Yalow RS, Glick SM, Roth J, ... Semax, a synthetic analogue fragment of adrenocorticotropic hormone Morton IK, Hall JM (December 6, 2012). Concise Dictionary ...
... (ACTH deficiency) is a result of a decreased or absent production of adrenocorticotropic ... hormone (ACTH) by the pituitary gland. It can be associated with TBX19. Symptoms include weakness, hypoglycemia, weight loss ...
Adrenocorticotropic+Hormone at the U.S. National Library of Medicine Medical Subject Headings (MeSH) Cosyntropin at the U.S. ... Adrenocorticotropic hormone is used as a medication and as diagnostic agent in the ACTH stimulation test.: 316, 1165 : 84, 271 ... March 2011). "Treatment of nephrotic syndrome with adrenocorticotropic hormone (ACTH) gel". Drug Design, Development and ... Both versions of the hormone are also used to perform the ACTH stimulation test to diagnose hypoadrenocorticism in dogs and ...
ISBN 978-0-07-142280-2. Schimmer BP, Parker KL (2006). "Adrenocorticotropic Hormone; Adrenocortical Steroids and Their ... July 2018). "Targeting hypothalamic-pituitary-adrenal axis hormones and sex steroids for improving cognition in major mood ... with a correlative assessment of androgen-related hormones". BJU Int. 101 (9): 1084-9. doi:10.1111/j.1464-410X.2008.07509.x. ... is a synthetic estrane steroid and a derivative of steroid hormones like progesterone, cortisol, and testosterone. It has ...
Takahashi A (2016). "Adrenocorticotropic Hormone". Handbook of Hormones. Elsevier. pp. 118-e16A-2. doi:10.1016/B978-0-12-801028 ... Corticotropes produce and release ACTH, a 39 amino acid peptide hormone, in response to corticotropic releasing hormone (CRH) ... and melanocyte-stimulating hormone (MSH). These cells are stimulated by corticotropin releasing hormone (CRH) and make up 15-20 ... These peptide hormones are stored within vesicles in the corticotropic cells and are released in response to CRH stimulation ...
DAX1 Adrenocorticotropic hormone deficiency; 201400; TBS19 Adrenoleukodystrophy; 300100; ABCD1 Adrenoleukodystrophy, neonatal; ... SECISBP2 Thyroid hormone resistance; 188570; THRB Thyroid hormone resistance, autosomal recessive; 274300; THRB Thyroid hormone ... HESX1 Growth hormone deficiency, isolated, type IA; 262400; GH1 Growth hormone deficiency, isolated, type IB; 612781; GH1 ... GHRHR Growth hormone deficiency, isolated, type II; 173100; GH1 Growth hormone insensitivity with immunodeficiency; 245590; ...
"Adrenocorticotropic hormone-independent Cushing's syndrome". Current Opinion in Endocrinology, Diabetes and Obesity. 14 (3): ... and zona reticularis that under normal conditions respond to body's physiological demands for steroid hormones. The adrenal ...
The adrenocorticotropic hormone receptor or ACTH receptor also known as the melanocortin receptor 2 or MC2 receptor is a type ... "ACTH - Clinical: Adrenocorticotropic Hormone (ACTH), Plasma". www.mayomedicallaboratories.com. Retrieved 2016-10-25. Gallo- ... adrenocorticotropic hormone) and melanocortin-3 receptors to chromosomes 18p11.2 and 20q13.2-q13.3 by fluorescence in situ ... "cDNA cloning and sequence analysis of the bovine adrenocorticotropic hormone (ACTH) receptor". Biochimica et Biophysica Acta ( ...
... adrenocorticotropic hormone) -> adrenal cortex secretes various stress hormones (e.g., cortisol) -> stress hormones (30 ... One thing that the body does to combat stressors is to create stress hormones, which in turn create energy reservoirs that are ...
"Mosby's Diagnostic and Laboratory Test Reference 2nd ed.: Adrenocorticotropic hormone stimulation test": 17-18. {{cite journal ... a progestogen steroid hormone related to progesterone Luteinizing hormone - a pituitary hormone that stimulates sex hormone ... stress response by measuring the adrenal response to adrenocorticotropic hormone (ACTH; corticotropin) or another corticotropic ... If secondary adrenal insufficiency is diagnosed, the insulin tolerance test (ITT) or the CRH (corticotropin-releasing hormone) ...
CRH Regulates secretion of adrenocorticotropic hormone (ACTH). CRH is widely distributed in the brain and periphery CRH also ... HPA axis activity and cytokines are intrinsically intertwined: inflammatory cytokines stimulate adrenocorticotropic hormone ( ... Anti-inflammatory hormones that enhance the organism's response to a stressor. Prevent the overreaction of the body's own ... Release of corticotropin-releasing hormone (CRH) from the hypothalamus is influenced by stress. CRH is a major regulator of the ...
Krueger RJ, Orme-Johnson NR (August 1983). "Acute adrenocorticotropic hormone stimulation of adrenal corticosteroidogenesis". J ... Hormones that stimulate its production depend on the cell type and include luteinizing hormone (LH), ACTH and angiotensin II. ... Srivastava VK, Vijayan E, Hiney JK, Dees WL (October 2005). "Effect of ethanol on follicle stimulating hormone-induced ... including altered steroid hormone levels and fertility. Alcohol DEHP and DBP Permethrin and cypermethrin DES and arsenite BPA ...
Growth and Adrenocorticotropic Hormones". Hormones of the anterior pituitary gland. I. Growth and adrenocorticotropic hormones ... In collaboration with various co-workers, he isolated several protein hormones, including adrenocorticotropic hormone (ACTH), ... The first group includes adrenocorticotropic hormone (ACTH), melanocyte-stimulating hormones (MSH), and lipotropin. 1953 - Li ... He isolated another pituitary hormone called melanocyte-stimulating hormone (MSH) and found that not only does this hormone ...
It comprises corticotropin-releasing factor (CRF), released by the hypothalamus; adrenocorticotropic hormone (ACTH), released ... the anterior pituitary hormone thyroid-stimulating hormone (TSH); and the thyroid hormones T3 and T4. •The hypothalamic- ... Two hormones are classically considered as being related to the posterior pituitary: oxytocin and vasopressin. These hormones ... the anterior pituitary luteinizing hormone (LH) and follicle-stimulating hormone (FSH), and the gonadal steroids. Hatton, GI ( ...
It comprises corticotropin-releasing factor (CRF), released by the hypothalamus; adrenocorticotropic hormone (ACTH), released ... the anterior pituitary hormone thyroid-stimulating hormone (TSH); and the thyroid hormones T3 and T4. •The hypothalamic- ... the anterior pituitary luteinizing hormone (LH) and follicle-stimulating hormone (FSH), and the gonadal steroids. Addis, B. J ... control the secretion of pituitary hormones, while others (the hormones oxytocin and vasopressin) are released directly into ...
The anterior pituitary in turn releases adrenocorticotropic hormone (ACTH). ACTH induces the release of corticosteroids and ... Stress hormones influence the processes carried out in the hippocampus and amygdala which are also associated with emotional ... These stress hormones are also hindering the hippocampus from receiving enough energy by diverting glucose levels to ... The varying effects of stress on performance or stress hormones are often compared to or known as "inverted-u" which induce ...
The trigger for this process in white adipose tissue is instead now thought to be adrenocorticotropic hormone, adrenaline and ... Kiwaki K, Levine JA (November 2003). "Differential effects of adrenocorticotropic hormone on human and mouse adipose tissue". ... The hormone leptin is primarily manufactured in the adipocytes of white adipose tissue which also produces another hormone, ... They have receptors for insulin, sex hormones, norepinephrine, and glucocorticoids. White adipose tissue is used for energy ...
... "hormone-sensitive lipase". Other catecholamines and adrenocorticotropic hormone (ACTH) can also stimulate such responses. Such ... Hormone stimulation of lipolysis in humans is similar to rats. The main function of hormone-sensitive lipase is to mobilize ... Extracellular hormones, such as glucagon, epinephrine, Thyroid-Stimulating Hormone, or ACTH, bind to their respective G protein ... "Entrez Gene: LIPE lipase, hormone-sensitive". Kraemer FB, Shen WJ (October 2002). "Hormone-sensitive lipase: control of ...
Two of these are adrenocorticotropic hormone (ACTH) and follicle stimulating hormone. The physiological role for responses to ... A particularly important bone-targeted hormonal regulator is parathyroid hormone (PTH). Parathyroid hormone is a protein made ... The skeleton is also modified for reproduction and in response to nutritional and other hormone stresses; it responds to ... Nicks KM, Fowler TW, Gaddy D (June 2010). "Reproductive hormones and bone". Curr Osteoporos Rep. 8 (2): 60-7. doi:10.1007/ ...
Urocortin acts in vitro to stimulate the secretion of adrenocorticotropic hormone. Urotensin is found in the teleost caudal ... The paraventricular nucleus transports CRH to the anterior pituitary, stimulating adrenocorticotropic hormone (ACTH) release ... Corticotropin-releasing hormone (CRH) is a releasing hormone found mainly in the paraventricular nucleus of the mammalian ... This family includes corticotropin-releasing hormone (also known as CRF), urotensin-I, urocortin, and sauvagine. The family can ...
It may worsen edema when taken alongside corticosteroids or adrenocorticotropic hormone. Like other AASs, oxandrolone is an ... However, a 2019 systematic review comparing effects of adding oxandrolone to growth hormone treatment to growth hormone alone ... When the same review assessed the effects of adding Oxandrolone to growth hormone treatment on speech, cognition and ... 2014). "Safety and efficacy of oxandrolone in growth hormone-treated girls with Turner syndrome: evidence from recent studies ...
The secretion of aldosterone is also stimulated by adrenocorticotropic hormone (ACTH). The cells of the zona glomerulosa do not ... The major hormone that stimulates cortisol secretion in humans is the ACTH that is released from the anterior pituitary. It has ... The adrenal cortex comprises three main zones, or layers that are regulated by distinct hormones as noted below. This anatomic ... Its secretion is regulated by the hormone ACTH from the anterior pituitary.[citation needed] They are produced mainly in the ...
Conversely, hydrocodone inhibits adrenocorticotropic hormone (ACTH), cortisol and luteinizing hormone (LH) secretion. Chronic ... Hydrocodone, like other opioids, stimulates the secretion of prolactin, growth hormone (GH), insulin and glucagon. ...
Raikhinstein M, Zohar M, Hanukoglu I (Feb 1994). "cDNA cloning and sequence analysis of the bovine adrenocorticotropic hormone ... Hanukoglu I (Dec 1992). "Steroidogenic enzymes: structure, function, and role in regulation of steroid hormone biosynthesis". ... in regulating steroid hormone synthesis in the adrenal cortex, regulation of adrenal steroidogenic capacity in disease states, ... Israel isolated the mitochondrial enzymes that catalyze the first step in the synthesis of steroid hormones in all ...
... adrenocorticotropic hormone (ACTH) and β-lipotropin. The formation of β-endorphin is then the result of cleavage of the C- ... However, POMC also gives rise to other peptide hormones, including α- and γ-melanocyte-stimulating hormone (MSH), resulting ... β-Endorphin (beta-endorphin) is an endogenous opioid neuropeptide and peptide hormone that is produced in certain neurons ... β-endorphin and other enkephalins are often released with ACTH to modulate hormone system functioning. Neuroregulation by β- ...
An adrenocorticotropic hormone stimulation test can be performed to assess adrenal function. Routine vaccination against ...
β-Melanocyte-stimulating hormone γ-Melanocyte-stimulating hormone Adrenocorticotropic hormone Varga, B.; Gesztelyi, R.; Bombicz ... α-Melanocyte-stimulating hormone (α-MSH) is an endogenous peptide hormone and neuropeptide of the melanocortin family, with a ... which is exclusive for adrenocorticotropic hormone (ACTH)). Activation of the MC1 receptor is responsible for its effect on ... It is the most important of the melanocyte-stimulating hormones (MSHs) (also known as melanotropins) in stimulating ...
These hormones are prolactin, growth hormone, TSH, adrenocorticotropic hormone, FSH and LH. They are all released by anterior ... Hormones of the hypothalamus, Hormones of the pituitary gland, Hormones of the hypothalamus-pituitary axis). ... Growth hormone releasing hormone (GHRH): stimulates GH secretion · Somatostatin: inhibits GH (and other hormone) secretion · ... Hypothalamic-pituitary hormones are hormones that are produced by the hypothalamus and pituitary gland. Although the organs in ...
Cortisol is important in signalling inhibition of adrenocorticotropic hormone release from the pituitary. Reduced cortisol in ... They do so by inhibiting the release of gonadotropin and luteinizing hormone, both hormones in the pituitary, responsible for ... activating the anterior pituitary and signalling the release of Adrenocorticotropic Hormone (ACTH), stimulating the adrenal ... Androgen is a steroid hormone, generally associated with development of male sex organs and secondary male sex characteristics ...
... stimulates the thyroid gland to make and release thyroid hormone.: 718 Adrenocorticotropic hormone (ACTH or corticotropin) - ... Tropic hormones are hormones that have other endocrine glands as their target. Most tropic hormones are produced and secreted ... Tropic hormones are contrasted with non-tropic hormones, which directly stimulate target cells. Tropic hormones from the ... 720-721 Endocrine system Non-tropic hormone Trophic hormone Purves, William K.; David Sadava; Gordon H. Orians; H. Craig Heller ...
Adrenocorticotropic hormone (ACTH; also adrenocorticotropin, corticotropin) is a polypeptide tropic hormone produced by and ... Adrenocorticotropic+Hormone at the U.S. National Library of Medicine Medical Subject Headings (MeSH) (Articles with short ... "Adrenocorticotropic Hormone (ACTH)". "Pro-opiomelocortin precursor". Retrieved April 8, 2013. Yalow RS, Glick SM, Roth J, ... Semax, a synthetic analogue fragment of adrenocorticotropic hormone Morton IK, Hall JM (December 6, 2012). Concise Dictionary ...
Collect blood into a plastic EDTA blood collection tube. Centrifuge and immediately transfer the plasma into a plastic shipping vial. Label sample as plasma. Freeze immediately and ship with freezer packs. Use of glass collection tubes may falsely decrease results ...
An adrenocorticotropic hormone (ACTH) test measures the amount of ACTH in your blood. It can help find the cause of abnormal ... What is an adrenocorticotropic hormone (ACTH) test?. This test measures the level of adrenocorticotropic hormone (ACTH) in a ... medlineplus.gov/lab-tests/adrenocorticotropic-hormone-acth/ Adrenocorticotropic Hormone (ACTH). ... Adrenocorticotropic Hormone (ACTH) Test; [modified 2022 Dec 14; cited 2023 May 23]; [about 11 screens]. Available from: https ...
A Novel Mechanism of Regulating Adrenocorticotropic Hormone Secretion? Message Subject (Your Name) has forwarded a page to you ... A Novel Mechanism of Regulating Adrenocorticotropic Hormone Secretion?. Carsten T. Wotjak, Masaharu Kubota, Gudrun Liebsch, ... A Novel Mechanism of Regulating Adrenocorticotropic Hormone Secretion?. Carsten T. Wotjak, Masaharu Kubota, Gudrun Liebsch, ... A Novel Mechanism of Regulating Adrenocorticotropic Hormone Secretion?. Carsten T. Wotjak, Masaharu Kubota, Gudrun Liebsch, ...
Adrenocorticotropic Hormone 1-24, Human CAS 16960-16-0 - Find MSDS or SDS, a COA, data sheets and more information. ... Synthetic, human adrenocorticotropic hormone (amino acids 1-24). N-Terminal synthetic fragment of pituitary hormone that ... Synthetic, human adrenocorticotropic hormone (amino acids 1-24). N-terminal synthetic fragment of the pituitary hormone, ... Adrenocorticotropic Hormone 1-24, Human MSDS (material safety data sheet) or SDS, CoA and CoQ, dossiers, brochures and other ...
This short article displays Pictures Of Adrenocorticotropic Hormone. Please click on the image(s) to view larger version. Feel ... Best viewed on 1280 x 768 px resolution in any modern browser.This article is about Pictures Of Adrenocorticotropic Hormone. ... This article is about Pictures Of Adrenocorticotropic Hormone.. All images are subject to our Image Copyright policy. If any of ... This short article displays Pictures Of Adrenocorticotropic Hormone. Please click on the image(s) to view larger version. Feel ...
Human ACTH(Adrenocorticotropic Hormone) ELISA Kit. Human ACTH(Adrenocorticotropic Hormone) ELISA Kit ... Description: This Rat Adrenocorticotropic hormone (ACTH) ELISA Kit employs a two-site sandwich ELISA to quantitate ACTH. ... Description: This Rat Adrenocorticotropic hormone (ACTH) ELISA Kit employs a two-site sandwich ELISA to quantitate ACTH. ... Description: A competitive Inhibition ELISA kit for detection of Adrenocorticotropic Hormone from Human in samples from blood, ...
Hormone Deficiency Symptoms. Adrenocorticotropic hormone deficiency. A deficiency of adrenocorticotropic hormone (ACTH), or ... Growth hormone deficiency. In children, growth hormone (GH) deficiency presents as growth retardation and delayed sexual ... Antidiuretic hormone deficiency. Antidiuretic hormone (ADH) deficiency causes polyuria and polydipsia (diabetes insipidus). ... Low levels of the gonadotropins--follicle-stimulating hormone (FSH) and luteinizing hormone (LH) --increase the risk of ...
Adrenocorticotropic Hormone (and Challenge Tests) answers are found in the Daviss Lab & Diagnostic Tests powered by Unbound ... "Adrenocorticotropic Hormone (and Challenge Tests)." Daviss Lab & Diagnostic Tests, 7th ed., F.A. Davis Company, 2017. Nursing ... Adrenocorticotropic Hormone (and Challenge Tests) [Internet]. In: Daviss Lab & Diagnostic Tests. F.A. Davis Company; 2017. [ ... TY - ELEC T1 - Adrenocorticotropic Hormone (and Challenge Tests) ID - 425331 A1 - Bladh,Mickey Lynn, AU - Van Leeuwen,Anne M, ...
Adrenocorticotropic Hormone human. Synonym(s). : Adrenocorticotropic hormone fragment 1-39, Corticotropin A ...
This report describes a case of Cushing syndrome due to ectopic adrenocorticotropic hor ... Disseminated Nocardiosis in Ectopic Adrenocorticotropic Hormone Syndrome: A Case Report : The Endocrinologist. ... Disseminated Nocardiosis in Ectopic Adrenocorticotropic Hormone Syndrome. A Case Report. Chrysanthidis, Theofilos MD*; ... This report describes a case of Cushing syndrome due to ectopic adrenocorticotropic hormone (ACTH) secretion, complicated with ...
Weberpals J, Djordjevic B, Khalifa M, Oza A. A rare case of ectopic adrenocorticotropic hormone syndrome in small cell ... A rare case of ectopic adrenocorticotropic hormone syndrome in small cell carcinoma of the vagina : A case report. In: Journal ... Weberpals, J., Djordjevic, B., Khalifa, M., & Oza, A. (2008). A rare case of ectopic adrenocorticotropic hormone syndrome in ... Weberpals, J, Djordjevic, B, Khalifa, M & Oza, A 2008, A rare case of ectopic adrenocorticotropic hormone syndrome in small ...
Adrenocorticotropic hormone *Headache. *Loss of vision. *Weight gain particularly on the face, neck, and trunk ... Depending on the type of hormone being produced, signs and symptoms of a functional pancreatic NET include:. *Gastrin * ... Gastrinoma: A tumor that forms in the cells that produce gastrin, a hormone responsible for making the stomach release acid to ... Glucagonoma: A tumor that forms in the cells that make glucagon, a hormone responsible for increasing the amount of glucose in ...
Adrenocorticotropic hormone. AR:. Androgen receptor. DHEAS:. Dehydroepiandrosterone sulfate. DHT:. Dihydrotestosterone. FGF:. ... The human skin as a hormone target and an endocrine gland. Hormones. 2004;3:9-26. ... Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans ... Growth hormone and insulin-like growth factors have different effects on sebaceous cell growth and differentiation. ...
... and adrenocorticotropic hormone (ACTH). No response in lung function was observed for 6 h after the LPS inhalation. The count ...
Adrenocorticotropic Hormone / blood * Adult * Animals * Arousal / physiology * Central Nervous System Stimulants * Circadian ...
... luteinizing hormone (LH), follicle stimulating hormone (FSH) and cortisol in stallions. Eight light horse and two pony ... administration of adrenocorticotropic hormone (ACTH) results in a testis-dependent, short-term increase in concentrations of ... Effect of administration of adrenocorticotropic hormone on plasma concentrations of testosterone, luteinizing hormone, follicle ... Effect of administration of adrenocorticotropic hormone on plasma concentrations of testosterone, luteinizing hormone, follicle ...
Impact of corticotropin-releasing hormone on gastrointestinal motility and adrenocorticotropic hormone in normal controls and ... Impact of corticotropin-releasing hormone on gastrointestinal motility and adrenocorticotropic hormone in normal controls and ... T1 - Impact of corticotropin-releasing hormone on gastrointestinal motility and adrenocorticotropic hormone in normal controls ... title = "Impact of corticotropin-releasing hormone on gastrointestinal motility and adrenocorticotropic hormone in normal ...
QY Research Medical published a comprehensive research on Adrenocorticotropic Hormone Receptor Market, which provides a ... Global Adrenocorticotropic Hormone Receptor market provides, wherever applicable and relevant, technical data of products, and ... Adrenocorticotropic Hormone Receptor Breakdown Data by Type. AQB-565. BIM-23B065. Corticotropin. Others ... The report scope of Adrenocorticotropic Hormone Receptor market covers the in-depth analysis, along with an overview of market ...
Adrenocorticotropic Hormone and PI3K/Akt Inhibition Reduce eNOS Phosphorylation and Increase Cortisol Biosynthesis in Long-Term ... and adrenocorticotropic hormone (ACTH) stimulation. UO126 reduced ACTH-stimulated cortisol in both normoxic and LTH FACs. UO126 ...
Adrenocorticotropic hormone (ACTH) / Cortisol. *DHEA-S. *Catecholamines (epinephrine, norepinephrine, dopamine). * ... Knowledge gained should include the peri-operative management of hormones, including insulin, octreotide, and thyroid hormone. ...
Inappropriate antidiuretic hormone secretion.. *Cushing syndrome from secretion of adrenocorticotropic hormone.. * ...
Plasma adrenocorticotropic hormone. Plasma ACTH was measured at time 0 (normal range 2-11 pmol·L−1) using a radioimmunoassay ( ... Sex-hormone analyses (at time 0): plasma follicle stimulating hormone (detection limit 0.05 IU·L−1), luteinizing hormone ( ... Adrenocorticotropic hormone test. Plasma cortisol was measured before (time 0), 30 and 60 min after intravenous (i.v.) ... Group 4: The adrenocorticotropic hormone (ACTH) test results from a group of 30 CF patients were retrospectively included as ...
Adrenocorticotropic hormone: adrenocortical steroids and their synthetic analogs; inhibitors of the synthesis and actions of ... adrenocortical hormones. In: Goodman LS, Gilman AG, eds. Goodman and Gilmans The Pharmacological Basis of Therapeutics. 8th ed ...
... and adrenocorticotropic hormone (ACTH) deficiency. (7) In the blood only 1 to 15% of cortisol is in its unbound or biologically ... Bovine hormones normally present in dairy products can cross-react with anti-cortisol antibodies and cause false results.. ... Hormone based oral contraceptives and estrogens can cause temporary increase in CBG, potentially lowering cortisol levels in ... Vining, R.F., McGinley, R.A., Symons, R.G. (1983). Hormones in saliva: mode of entry and consequent implications for clinical ...
Cushings disease, a type of Cushings syndrome in which an excessive amount of a hormone called adrenocorticotropic hormone ( ... Cortisol is a hormone made in the adrenal glands, which are small glands located near the top of each kidney. This hormone ... Testing measures the cortisol hormone in the blood, urine, or saliva. Cortisol is one of several glucocorticoid hormones that ... Ectopic Cushings syndrome, a condition in which the hormone ACTH is made in a tumor in another part of the body ...
3. Adrenocorticotropic Hormone ACTH.. 4. Alanine Transaminase (ALT, SGPT).. 5. Albumin, qualitative. ...
The ACTH test measures the level of adrenocorticotropic hormone (ACTH) in the blood. ACTH is a hormone released from the ... the more hormone in the bloodstream, the faster the chemical activity; the less hormone, the slower the activity). ... Endocrine glands release hormones (chemical messengers) into the bloodstream to be transported to various organs and tissues ... Pituitary gland not producing enough hormones, such as ACTH (hypopituitarism). *Tumor of the adrenal gland that produces too ...
  • ACTH is an important component of the hypothalamic-pituitary-adrenal axis and is often produced in response to biological stress (along with its precursor corticotropin-releasing hormone from the hypothalamus). (wikipedia.org)
  • POMC, ACTH and β-lipotropin are secreted from corticotropic cells in the anterior lobe (or adenohypophysis) of the pituitary gland in response to the hormone corticotropin-releasing hormone (CRH) released by the hypothalamus. (wikipedia.org)
  • ACTH consists of 39 amino acids, the first 13 of which (counting from the N-terminus) may be cleaved to form α-melanocyte-stimulating hormone (α-MSH) (this common structure is responsible for excessively tanned skin in Addison's disease). (wikipedia.org)
  • After a short period of time, ACTH is cleaved into α-melanocyte-stimulating hormone (α-MSH) and CLIP, a peptide with unknown activity in humans. (wikipedia.org)
  • ACTH stimulates secretion of glucocorticoid steroid hormones from adrenal cortex cells, especially in the zona fasciculata of the adrenal glands. (wikipedia.org)
  • Upon ligand binding, the receptor undergoes conformation changes that stimulate the enzyme adenylyl cyclase, which leads to an increase in intracellular cAMP and subsequent activation of protein kinase A. ACTH influences steroid hormone secretion by both rapid short-term mechanisms that take place within minutes and slower long-term actions. (wikipedia.org)
  • As indicated above, ACTH is a cleavage product of the pro-hormone, proopiomelanocortin (POMC), which also produces other hormones including α-MSH that stimulates the production of melanin. (wikipedia.org)
  • What is an adrenocorticotropic hormone (ACTH) test? (medlineplus.gov)
  • This test measures the level of adrenocorticotropic hormone (ACTH) in a sample of your blood. (medlineplus.gov)
  • ACTH tells your adrenal glands, two small glands that sit above your kidneys, to make another hormone called cortisol. (medlineplus.gov)
  • Your ACTH and cortisol levels are controlled by a complex feedback system of hormones made in different parts of your body. (medlineplus.gov)
  • It makes ACTH and other hormones. (medlineplus.gov)
  • It makes a hormone that tells your pituitary gland how much ACTH to make. (medlineplus.gov)
  • The increased amount of cortisol in your blood signals your hypothalamus to stop making the hormone that tells your pituitary to make ACTH. (medlineplus.gov)
  • Hypopituitarism , a condition in which the pituitary gland either stops making one or more hormones or can't make enough hormones, including ACTH. (medlineplus.gov)
  • This study evaluated adrenocorticotropic hormone (ACTH) responses in horses treated with pergolide and investigated factors that may influence response to treatment. (ivis.org)
  • Finally, a mixture of a V1 AVP and the α-helical corticotropin-releasing hormone (CRH) receptor antagonists administered via inverse microdialysis into the PVN caused a significant increase in the plasma adrenocorticotropic hormone (ACTH) concentration compared with vehicle-treated controls both under basal conditions and during social defeat, indicating inhibitory effects of intra-PVN-released AVP and/or CRH on HPA system activity. (jneurosci.org)
  • The hypothalamic-releasing factor, corticotropin-releasing hormone (CRH), stimulates the release of ACTH from the anterior pituitary gland. (unboundmedicine.com)
  • ACTH and cortisol are also measured together because cortisol and ACTH levels vary diurnally, with the peak values in both hormones occurring between 0600 and 0800 and reaching the lowest levels between 2200 and 2400. (unboundmedicine.com)
  • This report describes a case of Cushing syndrome due to ectopic adrenocorticotropic hormone (ACTH) secretion, complicated with disseminated nocardiosis. (lww.com)
  • To describe ectopic adrenocorticotropic hormone (ACTH) syndrome (EAS) to increase awareness of this condition when treating patients with gynecological small cell carcinoma (SmCC). (umn.edu)
  • Methods - CRH (2 χ/kg) was intravenously administered during duodenal and colonic manometry and plasma adrenocorticotropic hormone (ACTH) was measured by radioimmunoassay. (elsevierpure.com)
  • Cortisol production and peNOS were measured in response to pretreatment with the MEK /ERK1/2 pathway inhibitor UO126 (UO) and adrenocorticotropic hormone ( ACTH ) stimulation. (bvsalud.org)
  • An adrenocorticotrophic hormone (ACTH) test (250 µg tetracosactid) was performed in 25 CF patients treated with both itraconazole and budesonide, and in 12 patients treated with itraconazole alone (six patients with CF and six with chronic granulomateous disease). (ersjournals.com)
  • Mineralocorticoid and gonadal steroid function were evaluated by measurements of plasma-renin, follicle stimulating hormone, luteinising hormone, progesterone, oestradiol, testosterone, serum-inhibin A and B. ACTH tests performed as part of a pretransplantation programme in an additional 30 CF patients were used as controls. (ersjournals.com)
  • The ACTH test measures the level of adrenocorticotropic hormone (ACTH) in the blood. (mountsinai.org)
  • ACTH is a hormone released from the pituitary gland at the base of the brain. (mountsinai.org)
  • The main function of ACTH is to regulate the glucocorticoid (steroid) hormone cortisol. (mountsinai.org)
  • The adrenocorticotropic hormone (ACTH) response test serves to demonstrate functional adrenal reserve following administration of a pharmacological dose of ACTH. (vin.com)
  • The pituitary gland produces adrenocorticotropic hormone (ACTH), which stimulates the adrenal glands to produce other hormones. (medicalnewstoday.com)
  • Because cortisol synthesis is decreased, adrenocorticotropic hormone (ACTH) levels increase, which stimulates the adrenal cortex, causing accumulation of cortisol precursors (eg, 17- hydroxyprogesterone ) and excessive production of the adrenal androgens dehydroepiandrosterone (DHEA) and androstenedione. (merckmanuals.com)
  • Enzymes stimulated by adrenocorticotropic hormone (ACTH). (merckmanuals.com)
  • What type of hormone is ACTH? (kembrel.com)
  • ACTH) is a 39 amino acid peptide hormone produced by cells of the anterior pituitary gland and carried by the peripheral circulation to its effector organ, the adrenal cortex, where it stimulates the synthesis and secretion of glucocorticoids and, to a more modest extent. (kembrel.com)
  • A decline in the concentration of ACTH in the blood leads to a reduction in the secretion of adrenal hormones, resulting in adrenal insufficiency (hypoadrenalism). (kembrel.com)
  • What does the ACTH hormone do? (kembrel.com)
  • What is the normal range of ACTH hormone? (kembrel.com)
  • According to medical reports, the normal level of ACTH hormones is 6.0 to 76 pg/ml or 1.3 to 16.7 pmol/L. If the ACTH level of an individual is low compared to the normal value, then the person is suffering from Cushing syndrome. (kembrel.com)
  • ACTH is a hormone made by the pituitary gland, a small gland at the base of the brain. (kembrel.com)
  • ACTH controls the production of another hormone called cortisol. (kembrel.com)
  • Adrenocorticotropic hormone (ACTH), prolactin, cortisol, and heart rate responses decreased with repeated DMT administration, although blood pressure did not. (erowid.org)
  • Cortisol is a glucocorticoid (steroid) hormone released from the adrenal gland in response to adrenocorticotropic hormone (ACTH). (ucsfhealth.org)
  • In another study, eight healthy subjects were given dexamethasone 1 mg orally and tetracosactide [an adrenocorticotropic hormone (ACTH) analogue] 0.25 mg i.v., on separate occasions. (lu.se)
  • Release of Vasopressin within the Rat Paraventricular Nucleus in Response to Emotional Stress: A Novel Mechanism of Regulating Adrenocorticotropic Hormone Secretion? (jneurosci.org)
  • This causes a deficiency or loss of hypothalamic regulatory hormone input to the pituitary, resulting in loss of anterior pituitary hormone secretion. (medscape.com)
  • Inappropriate antidiuretic hormone secretion. (cancer.gov)
  • Cushing syndrome from secretion of adrenocorticotropic hormone. (cancer.gov)
  • The pathogenesis is most likely an itraconazole caused increase in systemic budesonide concentration through a reduced/inhibited metabolism leading to inhibition of adrenocorticotrophic hormone secretion along with a direct inhibition of steroidogenesis. (ersjournals.com)
  • More specifically, it stimulates secretion of glucocorticoids such as cortisol, and has little control over secretion of aldosterone, the other major steroid hormone from the adrenal cortex. (kembrel.com)
  • also adrenocorticotropin, corticotropin) is a polypeptide tropic hormone produced by and secreted by the anterior pituitary gland. (wikipedia.org)
  • Background - Corticotropin-releasing hormone (CRH) plays a key role in modulating intestinal motility in stressed animals. (elsevierpure.com)
  • An area of the brain called the hypothalamus produces the corticotropin-releasing hormone (CRH). (testing.com)
  • Yanovski JA, Cutler GB Jr, Chrousos GP, Nieman LK: The dexamethasone-suppressed corticotropin-releasing hormone stimulation test differentiates mild Cushing's disease from normal physiology. (karger.com)
  • End-organ hormonal insufficiencies are referred to as secondary deficiencies of the target organ (eg, hypothyroidism caused by a decrease in thyroid-stimulating hormone [TSH] is termed secondary hypothyroidism). (medscape.com)
  • The TSH (thyroid-stimulating hormone) test is a common blood test that measures the level of TSH in your bloodstream. (angis.org.au)
  • The TSH (thyroid-stimulating hormone) test is used to measure the level of TSH in your blood. (angis.org.au)
  • A TSH test, also known as a thyroid-stimulating hormone test, is a blood test that measures the level of TSH in your body. (angis.org.au)
  • Serum thyroid-stimulating hormone (TSH) and free thyroxine levels are within reference range in patients with classic Kallmann syndrome and idiopathic hypogonadotropic hypogonadism. (medscape.com)
  • Cortisol is one of several glucocorticoid hormones that help the body control blood sugar levels, respond to stress, and regulate the immune system. (testing.com)
  • Cortisol is a steroid (glucocorticoid or corticosteroid) hormone produced by the adrenal gland . (ucsfhealth.org)
  • The result is deficiency in some or all pituitary hormones. (medscape.com)
  • Growth hormone deficiency occurs when the pituitary gland does not produce enough growth hormone. (merckmanuals.com)
  • Growth hormone deficiency is the most common pituitary hormone deficiency and is accompanied by poor overall growth and short stature. (merckmanuals.com)
  • Other symptoms of growth hormone deficiency depend on the child's age and the cause of the deficiency. (merckmanuals.com)
  • Most often, doctors do not find a cause for growth hormone deficiency, but sometimes it is caused by a congenital disorder or brain tumor. (merckmanuals.com)
  • Hypopituitarism Hypopituitarism is an underactive pituitary gland that results in deficiency of one or more pituitary hormones. (merckmanuals.com)
  • In addition to a deficiency of growth hormone, short stature can occur for other reasons. (merckmanuals.com)
  • Prader-Willi syndrome (PWS) is a genetic multisystem disorder characterized during infancy by lethargy, diminished muscle tone (hypotonia), a weak suck and feeding difficulties with poor weight gain and growth and other hormone deficiency. (rarediseases.org)
  • Hypopituitarism is usually a combination of several hormonal deficiencies and rarely involves all pituitary hormones. (medscape.com)
  • Symptoms of hypopituitarism depend on what hormone is deficient and may include. (merckmanuals.com)
  • The pituitary, a pea-sized gland at the base of the brain, produces a number of hormones. (merckmanuals.com)
  • The medulla produces epinephrine-like hormones, while the cortex secretes corticosteroids . (medicalnewstoday.com)
  • Hypothyroidism occurs when the thyroid gland does not produce enough thyroid hormones, while hyperthyroidism occurs when the thyroid gland produces too much thyroid hormones. (angis.org.au)
  • One common reason is if you have symptoms of hypothyroidism, a condition where the thyroid gland doesn't produce enough hormones. (angis.org.au)
  • These hormones are produced in the hypothalamus but are stored in and released from the pituitary. (merckmanuals.com)
  • Adrenocorticotropic hormone, as its name implies, stimulates the adrenal cortex. (kembrel.com)
  • Damage to the cortex can disrupt the production of hormones such as cortisol, aldosterone, and androgens. (medicalnewstoday.com)
  • This is a frequently sampled serum luteinizing hormone (LH) profile in a male patient with Kallmann syndrome (KS) in comparison with a healthy individual. (medscape.com)
  • Testing measures the cortisol hormone in the blood, urine, or saliva. (testing.com)
  • What does cortisol hormone do? (kembrel.com)
  • A hormone is a chemical messenger in your bloodstream that controls the actions of certain cells or organs. (medlineplus.gov)
  • Endocrine glands release hormones (chemical messengers) into the bloodstream to be transported to various organs and tissues throughout the body. (mountsinai.org)
  • These hormones act on neighboring cells without entering the bloodstream. (proprofs.com)
  • These hormones act on neighboring cells without entering the bloodstream, which is a characteristic of both paracrines and autocrines. (proprofs.com)
  • Cortisol, the primary stress hormone, increases sugars (glucose) in the bloodstream, enhances your brain's use of glucose and increases the availability of substances that repair tissues. (kembrel.com)
  • In this paper, the potential role of gut hormones as potential treatments or predictors of response in depression is examined, with specific reference to the peptide hormone motilin. (mdpi.com)
  • Accumulated hormone precursors are shunted into androgen production, causing virilization. (merckmanuals.com)
  • Hormones are chemical messengers that affect the activity of another part of the body. (merckmanuals.com)
  • Examples of steroid hormones include cortisol, testosterone, and estrogen. (proprofs.com)
  • Adrenocorticotropic hormone directly stimulates testosterone production by the fetal and neonatal mouse testis. (kembrel.com)
  • Obesity decreases the sex hormone-binding globulin (SHBG) level and therefore decreases the total testosterone level. (medscape.com)
  • This hormone affects many processes in the body and influences the immune system, nervous system, and metabolism. (testing.com)
  • It is responsible for stimulating the thyroid gland to produce and release thyroid hormones, which regulate your body's metabolism. (angis.org.au)
  • Rare types of tumors of the cells that produce hormones responsible for controlling the balance of sugar , salt, and water in the body. (medicinenet.com)
  • They produce hormones that affect organs and tissues throughout the body. (medicalnewstoday.com)
  • In contrast, lipid soluble hormones, such as steroid hormones, can easily pass through the cell membrane due to their hydrophobic nature. (proprofs.com)
  • The signs or symptoms are either caused by the growth of the tumor and/or by the hormones produced by the tumor. (medicinenet.com)
  • TSH is produced by the pituitary gland and helps regulate the production of thyroid hormones. (angis.org.au)
  • A tumor that forms in the cells that make glucagon, a hormone responsible for increasing the amount of glucose in the blood. (medicinenet.com)
  • Although cortisol is the main hormone measured, other precursors can be assayed for investigation of sex hormone imbalance and non-cortisol producing tumours. (vin.com)
  • Diagnosis is by measurement of cortisol, its precursors, and adrenal androgens, sometimes after adrenocorticotropic hormone administration. (merckmanuals.com)
  • In the most common forms, accumulated hormone precursors. (merckmanuals.com)
  • If the pituitary gland does not produce enough growth hormone, abnormally slow growth and short stature can result. (merckmanuals.com)
  • Each of these hormones affects a specific part of the body (a target organ or tissue). (merckmanuals.com)
  • A tumor that forms in the cells that produce insulin, a hormone responsible for controlling the amount of glucose in the blood. (medicinenet.com)
  • To measure the amounts of glucose, potassium, and several hormone levels. (medicinenet.com)
  • An islet cell tumor, also known as a pancreatic neuroendocrine tumor , forms in hormone-producing cells (islet cells) of the pancreas. (medicinenet.com)
  • A tumor that forms in the cells that produce gastrin, a hormone responsible for making the stomach release acid to help digestion . (medicinenet.com)
  • Cortisol is a hormone made in the adrenal glands, which are small glands located near the top of each kidney. (testing.com)
  • Cortisol testing helps your doctor determine whether the adrenal glands are producing an appropriate amount of the hormone. (testing.com)
  • Addison's disease occurs when damage to the adrenal cortex disrupts hormone production in the adrenal glands. (medicalnewstoday.com)
  • Consequently, the adrenal glands will also produce lower levels of hormones. (medicalnewstoday.com)
  • It also plays a role in helping the body respond to stress and is sometimes called "the stress hormone. (testing.com)
  • Purpose Combining surgery and medical treatments allows the control of growth hormone hypersecretion in 80% of cases. (medscape.com)
  • Our objective was to determine the rate of acromegaly comorbidities once hypersecretion of growth hormone is controlled. (medscape.com)
  • Acromegaly is a rare disease generally due to a growth hormone (GH)-secreting pituitary adenoma. (medscape.com)