Wnt proteins are a large family of secreted glycoproteins that play essential roles in EMBRYONIC AND FETAL DEVELOPMENT, and tissue maintenance. They bind to FRIZZLED RECEPTORS and act as PARACRINE PROTEIN FACTORS to initiate a variety of SIGNAL TRANSDUCTION PATHWAYS. The canonical Wnt signaling pathway stabilizes the transcriptional coactivator BETA CATENIN.
A complex signaling pathway whose name is derived from the DROSOPHILA Wg gene, which when mutated results in the wingless phenotype, and the vertebrate INT gene, which is located near integration sites of MOUSE MAMMARY TUMOR VIRUS. The signaling pathway is initiated by the binding of WNT PROTEINS to cells surface WNT RECEPTORS which interact with the AXIN SIGNALING COMPLEX and an array of second messengers that influence the actions of BETA CATENIN.
A Wnt protein subtype that plays a role in cell-cell signaling during EMBRYONIC DEVELOPMENT and the morphogenesis of the developing NEURAL TUBE.
A Wnt protein subtype that plays a role in cell-cell signaling during EMBRYONIC DEVELOPMENT and the morphogenesis of the developing NEURAL TUBE. Defects in Wnt3 protein are associated with autosomal recessive tetra-AMELIA in humans.
A proto-oncogene protein and member of the Wnt family of proteins. It is expressed in the caudal MIDBRAIN and is essential for proper development of the entire mid-/hindbrain region.
A Wnt protein that is involved in regulating multiple developmental processes such as the formation of the KIDNEY; ADRENAL GLANDS; MAMMARY GLANDS; the PITUITARY GLAND; and the female reproductive system. Defects in WNT4 are a cause of ROKITANSKY KUSTER HAUSER SYNDROME.
A proto-oncogene protein and member of the Wnt family of proteins. It is frequently up-regulated in human GASTRIC CANCER and is a tumor marker (BIOMARKERS, TUMOR) of gastric and COLORECTAL CANCER.
Cell-surface receptors that specifically bind to WNT PROTEINS and activate the WNT SIGNALING PATHWAY.
A family of seven-pass transmembrane cell-surface proteins that combines with LOW DENSITY LIPROTEIN RECEPTOR-RELATED PROTEIN-5 or LOW DENSITY LIPROTEIN RECEPTOR-RELATED PROTEIN-5 to form receptors for WNT PROTEINS. Frizzled receptors often couple with HETEROTRIMERIC G PROTEINS and regulate the WNT SIGNALING PATHWAY.
The intracellular transfer of information (biological activation/inhibition) through a signal pathway. In each signal transduction system, an activation/inhibition signal from a biologically active molecule (hormone, neurotransmitter) is mediated via the coupling of a receptor/enzyme to a second messenger system or to an ion channel. Signal transduction plays an important role in activating cellular functions, cell differentiation, and cell proliferation. Examples of signal transduction systems are the GAMMA-AMINOBUTYRIC ACID-postsynaptic receptor-calcium ion channel system, the receptor-mediated T-cell activation pathway, and the receptor-mediated activation of phospholipases. Those coupled to membrane depolarization or intracellular release of calcium include the receptor-mediated activation of cytotoxic functions in granulocytes and the synaptic potentiation of protein kinase activation. Some signal transduction pathways may be part of larger signal transduction pathways; for example, protein kinase activation is part of the platelet activation signal pathway.
An LDL-receptor related protein that combines with cell surface FRIZZLED RECEPTORS to form WNT PROTEIN-binding receptors. The protein plays an important role in the WNT SIGNALING PATHWAY during EMBRYONIC DEVELOPMENT and in regulation of vascular cell proliferation.
Proteins obtained from the ZEBRAFISH. Many of the proteins in this species have been the subject of studies involving basic embryological development (EMBRYOLOGY).
A scaffolding protein that is a critical component of the axin signaling complex which binds to ADENOMATOUS POLYPOSIS COLI PROTEIN; GLYCOGEN SYNTHASE KINASE 3; and CASEIN KINASE I.
A family of DNA-binding proteins that are primarily expressed in T-LYMPHOCYTES. They interact with BETA CATENIN and serve as transcriptional activators and repressors in a variety of developmental processes.
Any of the processes by which nuclear, cytoplasmic, or intercellular factors influence the differential control of gene action during the developmental stages of an organism.
A family of proteins that share sequence similarity with the low density lipoprotein receptor (RECEPTORS, LDL).
A T-cell factor that plays an essential role in EMBRYONIC DEVELOPMENT.
Regulatory proteins and peptides that are signaling molecules involved in the process of PARACRINE COMMUNICATION. They are generally considered factors that are expressed by one cell and are responded to by receptors on another nearby cell. They are distinguished from HORMONES in that their actions are local rather than distal.
A family of cell surface receptors that were originally identified by their structural homology to neurotropic TYROSINE KINASES and referred to as orphan receptors because the associated ligand and signaling pathways were unknown. Evidence for the functionality of these proteins has been established by experiments showing that disruption of the orphan receptor genes results in developmental defects.
LDL-receptor related protein that combines with FRIZZLED RECEPTORS at the cell surface to form receptors that bind WNT PROTEINS. The protein plays an important role in the WNT SIGNALING PATHWAY in OSTEOBLASTS and during EMBRYONIC DEVELOPMENT.
Products of proto-oncogenes. Normally they do not have oncogenic or transforming properties, but are involved in the regulation or differentiation of cell growth. They often have protein kinase activity.
Proteins obtained from various species of Xenopus. Included here are proteins from the African clawed frog (XENOPUS LAEVIS). Many of these proteins have been the subject of scientific investigations in the area of MORPHOGENESIS and development.
The processes occurring in early development that direct morphogenesis. They specify the body plan ensuring that cells will proceed to differentiate, grow, and diversify in size and shape at the correct relative positions. Included are axial patterning, segmentation, compartment specification, limb position, organ boundary patterning, blood vessel patterning, etc.
Major constituent of the cytoskeleton found in the cytoplasm of eukaryotic cells. They form a flexible framework for the cell, provide attachment points for organelles and formed bodies, and make communication between parts of the cell possible.
The developmental entity of a fertilized egg (ZYGOTE) in animal species other than MAMMALS. For chickens, use CHICK EMBRYO.
An exotic species of the family CYPRINIDAE, originally from Asia, that has been introduced in North America. They are used in embryological studies and to study the effects of certain chemicals on development.
Progressive restriction of the developmental potential and increasing specialization of function that leads to the formation of specialized cells, tissues, and organs.
A technique that localizes specific nucleic acid sequences within intact chromosomes, eukaryotic cells, or bacterial cells through the use of specific nucleic acid-labeled probes.
A transcription factor that takes part in WNT signaling pathway. The activity of the protein is regulated via its interaction with BETA CATENIN. Transcription factor 7-like 2 protein plays an important role in the embryogenesis of the PANCREAS and ISLET CELLS.
The middle germ layer of an embryo derived from three paired mesenchymal aggregates along the neural tube.
Diffusible gene products that act on homologous or heterologous molecules of viral or cellular DNA to regulate the expression of proteins.
A TCF transcription factor that was originally identified as a DNA-binding protein that interacts with the enhancers of T-CELL RECEPTOR ALPHA GENES. It plays a role in T-LYMPHOCYTE development.
All of the processes involved in increasing CELL NUMBER including CELL DIVISION.
The largest family of cell surface receptors involved in SIGNAL TRANSDUCTION. They share a common structure and signal through HETEROTRIMERIC G-PROTEINS.
An aquatic genus of the family, Pipidae, occurring in Africa and distinguished by having black horny claws on three inner hind toes.
Endogenous substances, usually proteins, which are effective in the initiation, stimulation, or termination of the genetic transcription process.
A broad category of carrier proteins that play a role in SIGNAL TRANSDUCTION. They generally contain several modular domains, each of which having its own binding activity, and act by forming complexes with other intracellular-signaling molecules. Signal-transducing adaptor proteins lack enzyme activity, however their activity can be modulated by other signal-transducing enzymes
A process of complicated morphogenetic cell movements that reorganizes a bilayer embryo into one with three GERM LAYERS and specific orientation (dorsal/ventral; anterior/posterior). Gastrulation describes the germ layer development of a non-mammalian BLASTULA or that of a mammalian BLASTOCYST.
A negative regulator of beta-catenin signaling which is mutant in ADENOMATOUS POLYPOSIS COLI and GARDNER SYNDROME.
The development of anatomical structures to create the form of a single- or multi-cell organism. Morphogenesis provides form changes of a part, parts, or the whole organism.
Bone-growth regulatory factors that are members of the transforming growth factor-beta superfamily of proteins. They are synthesized as large precursor molecules which are cleaved by proteolytic enzymes. The active form can consist of a dimer of two identical proteins or a heterodimer of two related bone morphogenetic proteins.
Proteins encoded by homeobox genes (GENES, HOMEOBOX) that exhibit structural similarity to certain prokaryotic and eukaryotic DNA-binding proteins. Homeodomain proteins are involved in the control of gene expression during morphogenesis and development (GENE EXPRESSION REGULATION, DEVELOPMENTAL).
Laboratory mice that have been produced from a genetically manipulated EGG or EMBRYO, MAMMALIAN.
A cell line derived from cultured tumor cells.
Relatively undifferentiated cells that retain the ability to divide and proliferate throughout postnatal life to provide progenitor cells that can differentiate into specialized cells.
A salt of lithium that has been used experimentally as an immunomodulator.
A family of intercellular signaling proteins that play and important role in regulating the development of many TISSUES and organs. Their name derives from the observation of a hedgehog-like appearance in DROSOPHILA embryos with genetic mutations that block their action.
The developmental history of specific differentiated cell types as traced back to the original STEM CELLS in the embryo.
A cell line generated from human embryonic kidney cells that were transformed with human adenovirus type 5.
Established cell cultures that have the potential to propagate indefinitely.
Proteins which maintain the transcriptional quiescence of specific GENES or OPERONS. Classical repressor proteins are DNA-binding proteins that are normally bound to the OPERATOR REGION of an operon, or the ENHANCER SEQUENCES of a gene until a signal occurs that causes their release.
Theoretical representations that simulate the behavior or activity of biological processes or diseases. For disease models in living animals, DISEASE MODELS, ANIMAL is available. Biological models include the use of mathematical equations, computers, and other electronic equipment.
Any detectable and heritable change in the genetic material that causes a change in the GENOTYPE and which is transmitted to daughter cells and to succeeding generations.
A variation of the PCR technique in which cDNA is made from RNA via reverse transcription. The resultant cDNA is then amplified using standard PCR protocols.
The developmental stage that follows BLASTULA or BLASTOCYST. It is characterized by the morphogenetic cell movements including invagination, ingression, and involution. Gastrulation begins with the formation of the PRIMITIVE STREAK, and ends with the formation of three GERM LAYERS, the body plan of the mature organism.
Linear POLYPEPTIDES that are synthesized on RIBOSOMES and may be further modified, crosslinked, cleaved, or assembled into complex proteins with several subunits. The specific sequence of AMINO ACIDS determines the shape the polypeptide will take, during PROTEIN FOLDING, and the function of the protein.
The entity of a developing mammal (MAMMALS), generally from the cleavage of a ZYGOTE to the end of embryonic differentiation of basic structures. For the human embryo, this represents the first two months of intrauterine development preceding the stages of the FETUS.
Orientation of intracellular structures especially with respect to the apical and basolateral domains of the plasma membrane. Polarized cells must direct proteins from the Golgi apparatus to the appropriate domain since tight junctions prevent proteins from diffusing between the two domains.
Phosphoproteins are proteins that have been post-translationally modified with the addition of a phosphate group, usually on serine, threonine or tyrosine residues, which can play a role in their regulation, function, interaction with other molecules, and localization within the cell.
Conjugated protein-carbohydrate compounds including mucins, mucoid, and amyloid glycoproteins.
The complex processes of initiating CELL DIFFERENTIATION in the embryo. The precise regulation by cell interactions leads to diversity of cell types and specific pattern of organization (EMBRYOGENESIS).
The outer of the three germ layers of an embryo.
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.
Any of the processes by which nuclear, cytoplasmic, or intercellular factors influence the differential control of gene action in neoplastic tissue.
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.
Morphological and physiological development of EMBRYOS.
Bone-forming cells which secrete an EXTRACELLULAR MATRIX. HYDROXYAPATITE crystals are then deposited into the matrix to form bone.
ANIMALS whose GENOME has been altered by GENETIC ENGINEERING, or their offspring.
The artificial induction of GENE SILENCING by the use of RNA INTERFERENCE to reduce the expression of a specific gene. It includes the use of DOUBLE-STRANDED RNA, such as SMALL INTERFERING RNA and RNA containing HAIRPIN LOOP SEQUENCE, and ANTI-SENSE OLIGONUCLEOTIDES.

Axin prevents Wnt-3a-induced accumulation of beta-catenin. (1/399)

When Axin, a negative regulator of the Wnt signaling pathway, was expressed in COS cells, it coeluted with glycogen synthase kinase-3beta (GSK-3beta), beta-catenin, and adenomatous polyposis coli protein (APC) in a high molecular weight fraction on gel filtration column chromatography. In this fraction, GSK-3beta, beta-catenin, and APC were co-precipitated with Axin. Although beta-catenin was detected in the high molecular weight fraction in L cells on gel filtration column chromatography, addition of conditioned medium expressing Wnt-3a to the cells increased beta-catenin in the low molecular weight fraction. However, Wnt-3a-dependent accumulation of beta-catenin was greatly inhibited in L cells stably expressing Axin. Axin also suppressed Wnt-3a-dependent activation of Tcf-4 which binds to beta-catenin and acts as a transcription factor. These results suggest that Axin forms a complex with GSK-3beta, beta-catenin, and APC, resulting in the stimulation of the degradation of beta-catenin and that Wnt-3a induces the dissociation of beta-catenin from the Axin complex and accumulates beta-catenin.  (+info)

WNT signaling in the control of hair growth and structure. (2/399)

Characterization of the molecular pathways controlling differentiation and proliferation in mammalian hair follicles is central to our understanding of the regulation of normal hair growth, the basis of hereditary hair loss diseases, and the origin of follicle-based tumors. We demonstrate that the proto-oncogene Wnt3, which encodes a secreted paracrine signaling molecule, is expressed in developing and mature hair follicles and that its overexpression in transgenic mouse skin causes a short-hair phenotype due to altered differentiation of hair shaft precursor cells, and cyclical balding resulting from hair shaft structural defects and associated with an abnormal profile of protein expression in the hair shaft. A putative effector molecule for WNT3 signaling, the cytoplasmic protein Dishevelled 2 (DVL2), is normally present at high levels in a subset of cells in the outer root sheath and in precursor cells of the hair shaft cortex and cuticle which lie immediately adjacent to Wnt3-expressing cells. Overexpression of Dvl2 in the outer root sheath mimics the short-hair phenotype produced by overexpression of Wnt3, supporting the hypothesis that Wnt3 and Dvl2 have the potential to act in the same pathway in the regulation of hair growth. These experiments demonstrate a previously unrecognized role for WNT signaling in the control of hair growth and structure, as well as presenting the first example of a mammalian phenotype resulting from overexpression of a Dvl gene and providing an accessible in vivo system for analysis of mammalian WNT signaling pathways.  (+info)

Wnt3a-/--like phenotype and limb deficiency in Lef1(-/-)Tcf1(-/-) mice. (3/399)

Members of the LEF-1/TCF family of transcription factors have been implicated in the transduction of Wnt signals. However, targeted gene inactivations of Lef1, Tcf1, or Tcf4 in the mouse do not produce phenotypes that mimic any known Wnt mutation. Here we show that null mutations in both Lef1 and Tcf1, which are expressed in an overlapping pattern in the early mouse embryo, cause a severe defect in the differentiation of paraxial mesoderm and lead to the formation of additional neural tubes, phenotypes identical to those reported for Wnt3a-deficient mice. In addition, Lef1(-/-)Tcf1(-/-) embryos have defects in the formation of the placenta and in the development of limb buds, which fail both to express Fgf8 and to form an apical ectodermal ridge. Together, these data provide evidence for a redundant role of LEF-1 and TCF-1 in Wnt signaling during mouse development.  (+info)

Phosphorylation of axin, a Wnt signal negative regulator, by glycogen synthase kinase-3beta regulates its stability. (4/399)

Axin forms a complex with glycogen synthase kinase-3beta (GSK-3beta) and beta-catenin and promotes GSK-3beta-dependent phosphorylation of beta-catenin, thereby stimulating the degradation of beta-catenin. Because GSK-3beta also phosphorylates Axin in the complex, the physiological significance of the phosphorylation of Axin was examined. Treatment of COS cells with LiCl, a GSK-3beta inhibitor, and okadaic acid, a protein phosphatase inhibitor, decreased and increased, respectively, the cellular protein level of Axin. Pulse-chase analyses showed that the phosphorylated form of Axin was more stable than the unphosphorylated form and that an Axin mutant, in which the possible phosphorylation sites for GSK-3beta were mutated, exhibited a shorter half-life than wild type Axin. Dvl-1, which was genetically shown to function upstream of GSK-3beta, inhibited the phosphorylation of Axin by GSK-3beta in vitro. Furthermore, Wnt-3a-containing conditioned medium down-regulated Axin and accumulated beta-catenin in L cells and expression of Dvl-1(DeltaPDZ), in which the PDZ domain was deleted, suppressed this action of Wnt-3a. These results suggest that the phosphorylation of Axin is important for the regulation of its stability and that Wnt down-regulates Axin through Dvl.  (+info)

Retinoic acid induces down-regulation of Wnt-3a, apoptosis and diversion of tail bud cells to a neural fate in the mouse embryo. (5/399)

The tail bud comprises the caudal extremity of the vertebrate embryo, containing a pool of pluripotent mesenchymal stem cells that gives rise to almost all the tissues of the sacro-caudal region. Treatment of pregnant mice with 100 mg/kg all-trans retinoic acid at 9.5 days post coitum induces severe truncation of the body axis, providing a model system for studying the mechanisms underlying development of caudal agenesis. In the present study, we find that retinoic acid treatment causes extensive apoptosis of tail bud cells 24 h after treatment. Once the apoptotic cells have been removed, the remaining mesenchymal cells differentiate into an extensive network of ectopic tubules, radially arranged around the notochord. These tubules express Pax-3 and Pax-6 in a regionally-restricted pattern that closely resembles expression in the definitive neural tube. Neurofilament-positive neurons subsequently grow out from the ectopic tubules. Thus, the tail bud cells remaining after retinoic acid-induced apoptosis appear to adopt a neural fate. Wnt-3a, a gene that has been shown to be essential for tail bud formation, is specifically down-regulated in the tail bud of retinoic acid-treated embryos, as early as 2 h after retinoic acid treatment and Wnt-3a transcripts become undetectable by 10 h. In contrast, Wnt-5a and RAR-gamma are still detectable in the tail bud at that time. Extensive cell death also occurs in the tail bud of embryos homozygous for the vestigial tail mutation, in which there is a marked reduction in Wnt-3a expression. These embryos go on to develop multiple neural tubes in their truncated caudal region. These results suggest that retinoic acid induces down-regulation of Wnt-3a which may play an important role in the pathogenesis of axial truncation, involving induction of widespread apoptosis, followed by an alteration of tail bud cell fate to form multiple ectopic neural tubes.  (+info)

Human frizzled 1 interacts with transforming Wnts to transduce a TCF dependent transcriptional response. (6/399)

The human homologue of fz1 (Hfz1) was cloned from a cDNA library. Hfz1 was shown to couple to Wnt signal transduction pathways by its ability to enhance Wnt induced TCF dependent transcription in both autocrine and paracrine modes. Enhanced TCF dependent signaling was dose dependent with respect to both Wnt-3A and Hfz1. Moreover, Hfz1 deletion mutants with truncated carboxy termini showed markedly reduced capacity to enhance Wnt signal transduction. Specificity was demonstrated with respect to signal transduction by different Wnts. While Wnt-3a, -3, -1 and to a lesser extent Wnt-2 cooperated with Hfz1 in the paracrine assay for TCF dependent signaling, neither Wnt-4, -5a, -5b, -6, -7a nor -7b did so, despite similar levels of expression. However, coimmunoprecipitation of Hfz1 with both Wnt-3a and Wnt-5a indicated that TCF dependent signaling in response to Wnts is not determined solely by their ability to bind the receptor. All of these findings provide strong evidence that Hfz1 is a functional partner for certain Wnts in inducing TCF dependent transcription.  (+info)

T (Brachyury) is a direct target of Wnt3a during paraxial mesoderm specification. (7/399)

Wnt3a encodes a signal that is expressed in the primitive streak of the gastrulating mouse embryo and is required for paraxial mesoderm development. In its absence cells adopt ectopic neural fates. Embryos lacking the T-box-containing transcription factors, Brachyury or Tbx6, also lack paraxial mesoderm. Here we show that Brachyury is specifically down-regulated in Wnt3a mutants in cells fated to form paraxial mesoderm. Transgenic analysis of the T promoter identifies T (Brachyury) as a direct transcriptional target of the Wnt signaling pathway. Our results suggest that Wnt3a, signaling via Brachyury, modulates a balance between mesodermal and neural cell fates during gastrulation.  (+info)

A local Wnt-3a signal is required for development of the mammalian hippocampus. (8/399)

The mechanisms that regulate patterning and growth of the developing cerebral cortex remain unclear. Suggesting a role for Wnt signaling in these processes, multiple Wnt genes are expressed in selective patterns in the embryonic cortex. We have examined the role of Wnt-3a signaling at the caudomedial margin of the developing cerebral cortex, the site of hippocampal development. We show that Wnt-3a acts locally to regulate the expansion of the caudomedial cortex, from which the hippocampus develops. In mice lacking Wnt-3a, caudomedial cortical progenitor cells appear to be specified normally, but then underproliferate. By mid-gestation, the hippocampus is missing or represented by tiny populations of residual hippocampal cells. Thus, Wnt-3a signaling is crucial for the normal growth of the hippocampus. We suggest that the coordination of growth with patterning may be a general role for Wnts during vertebrate development.  (+info)

Wnt proteins are a family of secreted signaling molecules that play crucial roles in the regulation of fundamental biological processes, including cell proliferation, differentiation, migration, and survival. They were first discovered in 1982 through genetic studies in Drosophila melanogaster (fruit flies) and have since been found to be highly conserved across various species, from invertebrates to humans.

Wnt proteins exert their effects by binding to specific receptors on the target cell surface, leading to the activation of several intracellular signaling pathways:

1. Canonical Wnt/β-catenin pathway: In the absence of Wnt ligands, β-catenin is continuously degraded by a destruction complex consisting of Axin, APC (Adenomatous polyposis coli), and GSK3β (Glycogen synthase kinase 3 beta). When Wnt proteins bind to their receptors Frizzled and LRP5/6, the formation of a "signalosome" complex leads to the inhibition of the destruction complex, allowing β-catenin to accumulate in the cytoplasm and translocate into the nucleus. Here, it interacts with TCF/LEF (T-cell factor/lymphoid enhancer-binding factor) transcription factors to regulate the expression of target genes involved in cell proliferation, differentiation, and survival.
2. Non-canonical Wnt pathways: These include the Wnt/Ca^2+^ pathway and the planar cell polarity (PCP) pathway. In the Wnt/Ca^2+^ pathway, Wnt ligands bind to Frizzled receptors and activate heterotrimeric G proteins, leading to an increase in intracellular Ca^2+^ levels and activation of downstream targets such as protein kinase C (PKC) and calcium/calmodulin-dependent protein kinase II (CAMKII). These signaling events ultimately regulate cell movement, adhesion, and gene expression. In the PCP pathway, Wnt ligands bind to Frizzled receptors and coreceptor complexes containing Ror2 or Ryk, leading to activation of small GTPases such as RhoA and Rac1, which control cytoskeletal organization and cell polarity.

Dysregulation of Wnt signaling has been implicated in various human diseases, including cancer, developmental disorders, and degenerative conditions. In cancer, aberrant activation of the canonical Wnt/β-catenin pathway contributes to tumor initiation, progression, and metastasis by promoting cell proliferation, survival, and epithelial-mesenchymal transition (EMT). Inhibitors targeting different components of the Wnt signaling pathway are currently being developed as potential therapeutic strategies for cancer treatment.

The Wnt signaling pathway is a complex cell communication system that plays a critical role in embryonic development, tissue regeneration, and cancer. It is named after the Wingless (Wg) gene in Drosophila melanogaster and the Int-1 gene in mice, both of which were found to be involved in this pathway.

In essence, the Wnt signaling pathway involves the binding of Wnt proteins to Frizzled receptors on the cell surface, leading to the activation of intracellular signaling cascades. There are three main branches of the Wnt signaling pathway: the canonical (or Wnt/β-catenin) pathway, the noncanonical planar cell polarity (PCP) pathway, and the noncanonical Wnt/calcium pathway.

The canonical Wnt/β-catenin pathway is the most well-studied branch. In the absence of Wnt signaling, cytoplasmic β-catenin is constantly phosphorylated by a destruction complex consisting of Axin, APC, GSK3β, and CK1, leading to its ubiquitination and degradation in the proteasome. When Wnt ligands bind to Frizzled receptors and their coreceptor LRP5/6, Dishevelled is recruited and inhibits the destruction complex, allowing β-catenin to accumulate in the cytoplasm and translocate into the nucleus. In the nucleus, β-catenin interacts with TCF/LEF transcription factors to regulate the expression of target genes involved in cell proliferation, differentiation, and survival.

Dysregulation of the Wnt signaling pathway has been implicated in various human diseases, including cancer, developmental disorders, and degenerative conditions. For example, mutations in components of the canonical Wnt/β-catenin pathway can lead to the accumulation of β-catenin and subsequent activation of oncogenic target genes, contributing to tumorigenesis in various types of cancer.

Wnt3A is a type of Wnt protein, which is a secreted signaling molecule that plays crucial roles in the regulation of cell-to-cell communication during embryonic development and tissue homeostasis in adults. Specifically, Wnt3A is a member of the Wnt family that binds to Frizzled receptors and activates the canonical Wnt/β-catenin signaling pathway.

In this pathway, Wnt3A binding to its receptor leads to the inhibition of the β-catenin destruction complex, resulting in the stabilization and accumulation of β-catenin in the cytoplasm. β-catenin then translocates to the nucleus, where it interacts with TCF/LEF transcription factors to regulate the expression of target genes involved in cell proliferation, differentiation, and survival.

Wnt3A has been extensively studied in various biological contexts, including developmental biology, cancer research, and stem cell biology. In particular, Wnt3A has been shown to play important roles in the regulation of embryonic axis formation, neural crest development, and adult tissue regeneration. Dysregulation of Wnt/β-catenin signaling, including aberrant activation by Wnt3A, has been implicated in various human diseases, such as cancer, degenerative disorders, and fibrotic diseases.

Wnt3 protein is a member of the Wnt family of signaling proteins, which are secreted signaling molecules that play crucial roles in embryonic development and tissue homeostasis in adults. Specifically, Wnt3 is involved in the regulation of cell fate decisions, proliferation, and differentiation during embryogenesis. It binds to receptors on the target cells and activates a signaling pathway known as the canonical Wnt pathway, leading to the stabilization and nuclear accumulation of β-catenin, which then interacts with transcription factors to regulate gene expression. Defects in Wnt3 have been implicated in various developmental disorders, including some forms of congenital scoliosis and spina bifida.

Wnt1 protein is a member of the Wnt family, which is a group of secreted signaling proteins that play crucial roles in embryonic development and tissue homeostasis in adults. Specifically, Wnt1 is a highly conserved gene that encodes a glycoprotein with a molecular weight of approximately 40 kDa. It is primarily expressed in the developing nervous system, where it functions as a key regulator of neural crest cell migration and differentiation during embryogenesis.

Wnt1 protein mediates its effects by binding to Frizzled receptors on the surface of target cells, leading to the activation of several intracellular signaling pathways, including the canonical Wnt/β-catenin pathway and non-canonical Wnt/planar cell polarity (PCP) pathway. In the canonical pathway, Wnt1 protein stabilizes β-catenin, which then translocates to the nucleus and interacts with TCF/LEF transcription factors to regulate gene expression.

Dysregulation of Wnt1 signaling has been implicated in several human diseases, including cancer. For example, aberrant activation of the Wnt/β-catenin pathway by Wnt1 protein has been observed in various types of tumors, such as medulloblastomas and breast cancers, leading to uncontrolled cell proliferation and tumor growth. Therefore, understanding the molecular mechanisms underlying Wnt1 signaling is essential for developing novel therapeutic strategies for treating these diseases.

Wnt4 protein is a member of the Wnt family of signaling proteins, which are involved in various developmental processes, including cell fate determination, tissue homeostasis, and embryonic development. Specifically, Wnt4 plays crucial roles in female reproductive system development, such as promoting nephrogenesis (kidney development) and regulating Müllerian duct formation during sex differentiation. It exerts its functions by binding to Frizzled receptors and activating the canonical or non-canonical Wnt signaling pathways. Genetic mutations in WNT4 have been associated with certain genetic disorders, such as Mayer-Rokitansky-Küster-Hauser syndrome, which is characterized by congenital absence of the uterus and vagina.

Wnt2 protein is a member of the Wnt family of signaling proteins, which are involved in various developmental processes such as cell growth, division, and migration. Specifically, Wnt2 protein plays a role in the development of the nervous system and the immune system. It binds to receptors on the surface of cells and activates a signaling pathway that regulates gene expression. Dysregulation of Wnt2 protein has been implicated in several diseases, including cancer and neurodevelopmental disorders.

Wnt receptors are a group of cell surface proteins that play a crucial role in the Wnt signaling pathway. This pathway is essential for various biological processes during embryonic development and tissue regeneration in adults, including cell proliferation, differentiation, migration, and survival. The Wnt receptors can be broadly classified into two categories:

1. Frizzled (FZD) receptors: These are seven-transmembrane domain proteins that serve as the primary receptors for Wnt ligands. There are ten FZD receptor subtypes (FZD1-10) in humans, and they interact with different Wnt proteins to initiate distinct signaling cascades.
2. Low-density lipoprotein receptor-related protein (LRP) co-receptors: LRP5 and LRP6 are single-pass transmembrane proteins that function as co-receptors for Wnt ligands, typically in conjunction with FZD receptors. They contribute to the stabilization of the Wnt signaling complex and help amplify downstream signals.

The binding of a Wnt ligand to a FZD receptor and an LRP co-receptor triggers various intracellular signaling pathways, such as the canonical Wnt/β-catenin pathway or non-canonical Wnt pathways (e.g., Wnt/Ca^2+^ and planar cell polarity pathways). Dysregulation of Wnt receptors and their associated signaling pathways has been implicated in several diseases, including cancer, developmental disorders, and degenerative conditions.

Frizzled receptors are a type of cell surface receptor that are involved in the Wnt signaling pathway. They are named after the Drosophila melanogaster (fruit fly) mutant phenotype "frizzy" because of their role in regulating cell fate and patterning during development.

Frizzled receptors are composed of a seven-pass transmembrane domain, an extracellular cysteine-rich domain, and an intracellular tail. They bind to Wnt ligands, which are secreted proteins that play important roles in cell-cell communication during development and tissue homeostasis.

There are ten different Frizzled receptors identified in humans (FZD1-10) that can activate multiple signaling pathways, including the canonical Wnt/β-catenin pathway, noncanonical planar cell polarity pathway, and the Wnt/Ca2+ pathway. Dysregulation of Frizzled receptors has been implicated in various diseases, such as cancer, neurodevelopmental disorders, and metabolic disorders.

Signal transduction is the process by which a cell converts an extracellular signal, such as a hormone or neurotransmitter, into an intracellular response. This involves a series of molecular events that transmit the signal from the cell surface to the interior of the cell, ultimately resulting in changes in gene expression, protein activity, or metabolism.

The process typically begins with the binding of the extracellular signal to a receptor located on the cell membrane. This binding event activates the receptor, which then triggers a cascade of intracellular signaling molecules, such as second messengers, protein kinases, and ion channels. These molecules amplify and propagate the signal, ultimately leading to the activation or inhibition of specific cellular responses.

Signal transduction pathways are highly regulated and can be modulated by various factors, including other signaling molecules, post-translational modifications, and feedback mechanisms. Dysregulation of these pathways has been implicated in a variety of diseases, including cancer, diabetes, and neurological disorders.

Low-Density Lipoprotein Receptor-Related Protein 6 (LRP6) is a protein that is encoded by the LRP6 gene in humans. It is a member of the low-density lipoprotein receptor family and plays a crucial role in signal transduction pathways, particularly the Wnt signaling pathway.

In the Wnt signaling pathway, LRP6 acts as a co-receptor for Wnt proteins, which are involved in various developmental processes, including cell fate determination, proliferation, and migration. When Wnt proteins bind to LRP6 and other receptors, they trigger a cascade of intracellular signaling events that ultimately lead to the regulation of gene expression.

Mutations in the LRP6 gene have been associated with several human diseases, including familial exudative vitreoretinopathy, a genetic disorder that affects the eyes, and various forms of cancer. Additionally, abnormalities in LRP6 function have been implicated in the development of conditions such as Alzheimer's disease, diabetes, and metabolic disorders.

Zebrafish proteins refer to the diverse range of protein molecules that are produced by the organism Danio rerio, commonly known as the zebrafish. These proteins play crucial roles in various biological processes such as growth, development, reproduction, and response to environmental stimuli. They are involved in cellular functions like enzymatic reactions, signal transduction, structural support, and regulation of gene expression.

Zebrafish is a popular model organism in biomedical research due to its genetic similarity with humans, rapid development, and transparent embryos that allow for easy observation of biological processes. As a result, the study of zebrafish proteins has contributed significantly to our understanding of protein function, structure, and interaction in both zebrafish and human systems.

Some examples of zebrafish proteins include:

* Transcription factors that regulate gene expression during development
* Enzymes involved in metabolic pathways
* Structural proteins that provide support to cells and tissues
* Receptors and signaling molecules that mediate communication between cells
* Heat shock proteins that assist in protein folding and protect against stress

The analysis of zebrafish proteins can be performed using various techniques, including biochemical assays, mass spectrometry, protein crystallography, and computational modeling. These methods help researchers to identify, characterize, and understand the functions of individual proteins and their interactions within complex networks.

Axin protein is a type of intracellular protein that plays a crucial role in regulating the Wnt signaling pathway, which is essential for various developmental processes and tissue homeostasis. Axin serves as a scaffold protein that facilitates the formation of a complex with other proteins involved in the degradation of β-catenin, a key component of the Wnt signalling cascade. By promoting the phosphorylation and subsequent degradation of β-catenin, Axin helps to maintain its levels in the cell and ensures proper regulation of gene transcription. Mutations in the AXIN gene can lead to abnormal Wnt signaling and have been associated with various diseases, including cancer.

TCF (T-cell factor) transcription factors are a family of proteins that play a crucial role in the Wnt signaling pathway, which is involved in various biological processes such as cell proliferation, differentiation, and migration. TCF transcription factors bind to specific DNA sequences in the promoter region of target genes and regulate their transcription.

In the absence of Wnt signaling, TCF proteins form a complex with transcriptional repressors, which inhibits gene transcription. When Wnt ligands bind to their receptors, they initiate a cascade of intracellular signals that result in the accumulation and nuclear localization of β-catenin, a key player in the Wnt signaling pathway.

In the nucleus, β-catenin interacts with TCF proteins, displacing the transcriptional repressors and converting TCF into an activator of gene transcription. This leads to the expression of target genes that are involved in various cellular processes, including cell cycle regulation, stem cell maintenance, and tumorigenesis.

Mutations in TCF transcription factors or components of the Wnt signaling pathway have been implicated in several human diseases, including cancer, developmental disorders, and degenerative diseases.

Developmental gene expression regulation refers to the processes that control the activation or repression of specific genes during embryonic and fetal development. These regulatory mechanisms ensure that genes are expressed at the right time, in the right cells, and at appropriate levels to guide proper growth, differentiation, and morphogenesis of an organism.

Developmental gene expression regulation is a complex and dynamic process involving various molecular players, such as transcription factors, chromatin modifiers, non-coding RNAs, and signaling molecules. These regulators can interact with cis-regulatory elements, like enhancers and promoters, to fine-tune the spatiotemporal patterns of gene expression during development.

Dysregulation of developmental gene expression can lead to various congenital disorders and developmental abnormalities. Therefore, understanding the principles and mechanisms governing developmental gene expression regulation is crucial for uncovering the etiology of developmental diseases and devising potential therapeutic strategies.

LDL-Receptor Related Proteins (LRP) are a family of single transmembrane domain receptors that play important roles in various cellular processes, including endocytosis, intracellular signaling, and protein degradation. They are named after their structural and functional similarities to the low-density lipoprotein (LDL) receptor.

The LDL-Receptor Related Proteins consist of several members, including LRP1, LRP2 (also known as Megalin), LRP3, LRP4, LRP5, and LRP6. These proteins are widely expressed in various tissues, such as the brain, liver, kidney, and muscle.

LRP1 is a large receptor that is involved in the clearance of several ligands, including LDL, apolipoprotein E (apoE), and α2-macroglobulin. It also plays a role in intracellular signaling pathways related to cell survival, differentiation, and migration.

LRP2 is primarily expressed in the kidney and the brain, where it functions as a scavenger receptor that mediates the endocytosis of various ligands, including lipoproteins, vitamin-binding proteins, and enzymes.

LRP3 is involved in the clearance of apoE-containing lipoproteins and has been implicated in the regulation of cholesterol metabolism.

LRP4 is a critical regulator of neuromuscular junction formation and function, and it interacts with several ligands, including agrin and LDL.

LRP5 and LRP6 are involved in the Wnt signaling pathway, which plays important roles in embryonic development, tissue homeostasis, and cancer. They act as co-receptors for Wnt proteins and modulate intracellular signaling pathways that regulate gene expression and cell behavior.

Overall, LDL-Receptor Related Proteins play diverse and critical roles in various physiological processes, and their dysfunction has been implicated in several diseases, including neurodegenerative disorders, cardiovascular disease, and cancer.

Lymphoid Enhancer-Binding Factor 1 (LEF1) is a protein that functions as a transcription factor, playing a crucial role in the Wnt signaling pathway. It is involved in the regulation of gene expression, particularly during embryonic development and immune system function. LEF1 helps control the differentiation and proliferation of certain cells, including B and T lymphocytes, which are essential for adaptive immunity. Mutations in the LEF1 gene have been associated with various human diseases, such as cancer and immunodeficiency disorders.

Intercellular signaling peptides and proteins are molecules that mediate communication and interaction between different cells in living organisms. They play crucial roles in various biological processes, including cell growth, differentiation, migration, and apoptosis (programmed cell death). These signals can be released into the extracellular space, where they bind to specific receptors on the target cell's surface, triggering intracellular signaling cascades that ultimately lead to a response.

Peptides are short chains of amino acids, while proteins are larger molecules made up of one or more polypeptide chains. Both can function as intercellular signaling molecules by acting as ligands for cell surface receptors or by being cleaved from larger precursor proteins and released into the extracellular space. Examples of intercellular signaling peptides and proteins include growth factors, cytokines, chemokines, hormones, neurotransmitters, and their respective receptors.

These molecules contribute to maintaining homeostasis within an organism by coordinating cellular activities across tissues and organs. Dysregulation of intercellular signaling pathways has been implicated in various diseases, such as cancer, autoimmune disorders, and neurodegenerative conditions. Therefore, understanding the mechanisms underlying intercellular signaling is essential for developing targeted therapies to treat these disorders.

Receptor Tyrosine Kinase-like Orphan Receptors (RORs) are a subfamily of transmembrane receptors that share structural similarities with Receptor Tyrosine Kinases (RTKs), but their ligands and precise functions are not well understood. RORs have an extracellular domain, a single transmembrane region, and an intracellular kinase-like domain, which is thought to be catalytically inactive. There are two members of this subfamily, ROR1 and ROR2, which are involved in various developmental processes, including cell proliferation, differentiation, and migration. Because their physiological roles are not fully elucidated, they are referred to as "orphan receptors." However, recent research has uncovered potential ligands and signaling pathways for RORs, contributing to a better understanding of their functions in health and disease.

Low-Density Lipoprotein Receptor-Related Protein 5 (LRP5) is a gene that encodes a single transmembrane protein, which belongs to the low-density lipoprotein receptor family. This protein plays crucial roles in various biological processes such as bone formation, vascular biology, and cancer progression. It functions as a co-receptor for Wnt signaling pathways that regulate gene expression involved in cell proliferation, differentiation, and migration.

In the context of lipid metabolism, LRP5 has been shown to modulate lipoprotein particle uptake and cholesterol homeostasis through its interaction with LDL receptors. Mutations in the LRP5 gene have been associated with several diseases, including osteoporosis, familial exudative vitreoretinopathy, and adiposity-associated disorders.

In summary, Low-Density Lipoprotein Receptor-Related Protein 5 is a multifunctional receptor involved in various physiological processes, including bone formation, lipid metabolism, and cell signaling.

Proto-oncogene proteins are normal cellular proteins that play crucial roles in various cellular processes, such as signal transduction, cell cycle regulation, and apoptosis (programmed cell death). They are involved in the regulation of cell growth, differentiation, and survival under physiological conditions.

When proto-oncogene proteins undergo mutations or aberrations in their expression levels, they can transform into oncogenic forms, leading to uncontrolled cell growth and division. These altered proteins are then referred to as oncogene products or oncoproteins. Oncogenic mutations can occur due to various factors, including genetic predisposition, environmental exposures, and aging.

Examples of proto-oncogene proteins include:

1. Ras proteins: Involved in signal transduction pathways that regulate cell growth and differentiation. Activating mutations in Ras genes are found in various human cancers.
2. Myc proteins: Regulate gene expression related to cell cycle progression, apoptosis, and metabolism. Overexpression of Myc proteins is associated with several types of cancer.
3. EGFR (Epidermal Growth Factor Receptor): A transmembrane receptor tyrosine kinase that regulates cell proliferation, survival, and differentiation. Mutations or overexpression of EGFR are linked to various malignancies, such as lung cancer and glioblastoma.
4. Src family kinases: Intracellular tyrosine kinases that regulate signal transduction pathways involved in cell proliferation, survival, and migration. Dysregulation of Src family kinases is implicated in several types of cancer.
5. Abl kinases: Cytoplasmic tyrosine kinases that regulate various cellular processes, including cell growth, differentiation, and stress responses. Aberrant activation of Abl kinases, as seen in chronic myelogenous leukemia (CML), leads to uncontrolled cell proliferation.

Understanding the roles of proto-oncogene proteins and their dysregulation in cancer development is essential for developing targeted cancer therapies that aim to inhibit or modulate these aberrant signaling pathways.

"Xenopus proteins" refer to the proteins that are expressed or isolated from the Xenopus species, which are primarily used as model organisms in biological and biomedical research. The most commonly used Xenopus species for research are the African clawed frogs, Xenopus laevis and Xenopus tropicalis. These proteins play crucial roles in various cellular processes and functions, and they serve as valuable tools to study different aspects of molecular biology, developmental biology, genetics, and biochemistry.

Some examples of Xenopus proteins that are widely studied include:

1. Xenopus Histones: These are the proteins that package DNA into nucleosomes, which are the fundamental units of chromatin in eukaryotic cells. They play a significant role in gene regulation and epigenetic modifications.
2. Xenopus Cyclins and Cyclin-dependent kinases (CDKs): These proteins regulate the cell cycle and control cell division, differentiation, and apoptosis.
3. Xenopus Transcription factors: These proteins bind to specific DNA sequences and regulate gene expression during development and in response to various stimuli.
4. Xenopus Signaling molecules: These proteins are involved in intracellular signaling pathways that control various cellular processes, such as cell growth, differentiation, migration, and survival.
5. Xenopus Cytoskeletal proteins: These proteins provide structural support to the cells and regulate their shape, motility, and organization.
6. Xenopus Enzymes: These proteins catalyze various biochemical reactions in the cell, such as metabolic pathways, DNA replication, transcription, and translation.

Overall, Xenopus proteins are essential tools for understanding fundamental biological processes and have contributed significantly to our current knowledge of molecular biology, genetics, and developmental biology.

"Body patterning" is a general term that refers to the process of forming and organizing various tissues and structures into specific patterns during embryonic development. This complex process involves a variety of molecular mechanisms, including gene expression, cell signaling, and cell-cell interactions. It results in the creation of distinct body regions, such as the head, trunk, and limbs, as well as the organization of internal organs and systems.

In medical terminology, "body patterning" may refer to specific developmental processes or abnormalities related to embryonic development. For example, in genetic disorders such as Poland syndrome or Holt-Oram syndrome, mutations in certain genes can lead to abnormal body patterning, resulting in the absence or underdevelopment of certain muscles, bones, or other structures.

It's important to note that "body patterning" is not a formal medical term with a specific definition, but rather a general concept used in developmental biology and genetics.

Cytoskeletal proteins are a type of structural proteins that form the cytoskeleton, which is the internal framework of cells. The cytoskeleton provides shape, support, and structure to the cell, and plays important roles in cell division, intracellular transport, and maintenance of cell shape and integrity.

There are three main types of cytoskeletal proteins: actin filaments, intermediate filaments, and microtubules. Actin filaments are thin, rod-like structures that are involved in muscle contraction, cell motility, and cell division. Intermediate filaments are thicker than actin filaments and provide structural support to the cell. Microtubules are hollow tubes that are involved in intracellular transport, cell division, and maintenance of cell shape.

Cytoskeletal proteins are composed of different subunits that polymerize to form filamentous structures. These proteins can be dynamically assembled and disassembled, allowing cells to change their shape and move. Mutations in cytoskeletal proteins have been linked to various human diseases, including cancer, neurological disorders, and muscular dystrophies.

A nonmammalian embryo refers to the developing organism in animals other than mammals, from the fertilized egg (zygote) stage until hatching or birth. In nonmammalian species, the developmental stages and terminology differ from those used in mammals. The term "embryo" is generally applied to the developing organism up until a specific stage of development that is characterized by the formation of major organs and structures. After this point, the developing organism is referred to as a "larva," "juvenile," or other species-specific terminology.

The study of nonmammalian embryos has played an important role in our understanding of developmental biology and evolutionary developmental biology (evo-devo). By comparing the developmental processes across different animal groups, researchers can gain insights into the evolutionary origins and diversification of body plans and structures. Additionally, nonmammalian embryos are often used as model systems for studying basic biological processes, such as cell division, gene regulation, and pattern formation.

A zebrafish is a freshwater fish species belonging to the family Cyprinidae and the genus Danio. Its name is derived from its distinctive striped pattern that resembles a zebra's. Zebrafish are often used as model organisms in scientific research, particularly in developmental biology, genetics, and toxicology studies. They have a high fecundity rate, transparent embryos, and a rapid development process, making them an ideal choice for researchers. However, it is important to note that providing a medical definition for zebrafish may not be entirely accurate or relevant since they are primarily used in biological research rather than clinical medicine.

Cell differentiation is the process by which a less specialized cell, or stem cell, becomes a more specialized cell type with specific functions and structures. This process involves changes in gene expression, which are regulated by various intracellular signaling pathways and transcription factors. Differentiation results in the development of distinct cell types that make up tissues and organs in multicellular organisms. It is a crucial aspect of embryonic development, tissue repair, and maintenance of homeostasis in the body.

In situ hybridization (ISH) is a molecular biology technique used to detect and localize specific nucleic acid sequences, such as DNA or RNA, within cells or tissues. This technique involves the use of a labeled probe that is complementary to the target nucleic acid sequence. The probe can be labeled with various types of markers, including radioisotopes, fluorescent dyes, or enzymes.

During the ISH procedure, the labeled probe is hybridized to the target nucleic acid sequence in situ, meaning that the hybridization occurs within the intact cells or tissues. After washing away unbound probe, the location of the labeled probe can be visualized using various methods depending on the type of label used.

In situ hybridization has a wide range of applications in both research and diagnostic settings, including the detection of gene expression patterns, identification of viral infections, and diagnosis of genetic disorders.

Transcription Factor 7-Like 2 Protein (TF7L2) is a transcription factor that plays a crucial role in the Wnt signaling pathway, which is essential for cell differentiation, proliferation, and apoptosis. It is primarily expressed in the pancreas, brain, and muscle tissues.

TF7L2 is involved in the regulation of gene expression, particularly those related to insulin synthesis and secretion in the pancreatic beta-cells. Variations in the TF7L2 gene have been associated with an increased risk of developing type 2 diabetes, as they can affect insulin sensitivity and glucose metabolism.

Mutations in the TF7L2 gene may lead to abnormal regulation of genes involved in glucose homeostasis, which can contribute to impaired insulin secretion and the development of type 2 diabetes. However, the exact mechanisms by which TF7L2 variants increase the risk of type 2 diabetes are not fully understood and are an area of ongoing research.

In medical and embryological terms, the mesoderm is one of the three primary germ layers in the very early stages of embryonic development. It forms between the ectoderm and endoderm during gastrulation, and it gives rise to a wide variety of cell types, tissues, and organs in the developing embryo.

The mesoderm contributes to the formation of structures such as:

1. The connective tissues (including tendons, ligaments, and most of the bones)
2. Muscular system (skeletal, smooth, and cardiac muscles)
3. Circulatory system (heart, blood vessels, and blood cells)
4. Excretory system (kidneys and associated structures)
5. Reproductive system (gonads, including ovaries and testes)
6. Dermis of the skin
7. Parts of the eye and inner ear
8. Several organs in the urogenital system

Dysfunctions or abnormalities in mesoderm development can lead to various congenital disorders and birth defects, highlighting its importance during embryogenesis.

Trans-activators are proteins that increase the transcriptional activity of a gene or a set of genes. They do this by binding to specific DNA sequences and interacting with the transcription machinery, thereby enhancing the recruitment and assembly of the complexes needed for transcription. In some cases, trans-activators can also modulate the chromatin structure to make the template more accessible to the transcription machinery.

In the context of HIV (Human Immunodeficiency Virus) infection, the term "trans-activator" is often used specifically to refer to the Tat protein. The Tat protein is a viral regulatory protein that plays a critical role in the replication of HIV by activating the transcription of the viral genome. It does this by binding to a specific RNA structure called the Trans-Activation Response Element (TAR) located at the 5' end of all nascent HIV transcripts, and recruiting cellular cofactors that enhance the processivity and efficiency of RNA polymerase II, leading to increased viral gene expression.

T-cell transcription factor 1 (TFH1), also known as TCF7, is a protein that plays a crucial role in the development and function of T cells, which are a type of white blood cell involved in immune response. TFH1 is a transcription factor, meaning it binds to specific regions of DNA and helps control the expression of genes involved in T cell activation, differentiation, and survival.

TFH1 is part of a family of transcription factors called basic helix-loop-helix proteins, which are characterized by a conserved region known as the bHLH domain. This domain allows TFH1 to bind to DNA and regulate gene expression. In T cells, TFH1 helps control the expression of genes involved in T cell activation and differentiation, including those that encode cytokine receptors and other transcription factors.

Mutations in the gene that encodes TFH1 (TCF7) have been associated with various immune disorders, including autoimmune diseases and primary immunodeficiencies. Additionally, recent research has suggested that TFH1 may play a role in cancer biology, as it has been shown to be upregulated in certain types of tumors and may contribute to tumor growth and progression.

Cell proliferation is the process by which cells increase in number, typically through the process of cell division. In the context of biology and medicine, it refers to the reproduction of cells that makes up living tissue, allowing growth, maintenance, and repair. It involves several stages including the transition from a phase of quiescence (G0 phase) to an active phase (G1 phase), DNA replication in the S phase, and mitosis or M phase, where the cell divides into two daughter cells.

Abnormal or uncontrolled cell proliferation is a characteristic feature of many diseases, including cancer, where deregulated cell cycle control leads to excessive and unregulated growth of cells, forming tumors that can invade surrounding tissues and metastasize to distant sites in the body.

G-protein-coupled receptors (GPCRs) are a family of membrane receptors that play an essential role in cellular signaling and communication. These receptors possess seven transmembrane domains, forming a structure that spans the lipid bilayer of the cell membrane. They are called "G-protein-coupled" because they interact with heterotrimeric G proteins upon activation, which in turn modulate various downstream signaling pathways.

When an extracellular ligand binds to a GPCR, it causes a conformational change in the receptor's structure, leading to the exchange of guanosine diphosphate (GDP) for guanosine triphosphate (GTP) on the associated G protein's α subunit. This exchange triggers the dissociation of the G protein into its α and βγ subunits, which then interact with various effector proteins to elicit cellular responses.

There are four main families of GPCRs, classified based on their sequence similarities and downstream signaling pathways:

1. Gq-coupled receptors: These receptors activate phospholipase C (PLC), which leads to the production of inositol trisphosphate (IP3) and diacylglycerol (DAG). IP3 induces calcium release from intracellular stores, while DAG activates protein kinase C (PKC).
2. Gs-coupled receptors: These receptors activate adenylyl cyclase, which increases the production of cyclic adenosine monophosphate (cAMP) and subsequently activates protein kinase A (PKA).
3. Gi/o-coupled receptors: These receptors inhibit adenylyl cyclase, reducing cAMP levels and modulating PKA activity. Additionally, they can activate ion channels or regulate other signaling pathways through the βγ subunits.
4. G12/13-coupled receptors: These receptors primarily activate RhoGEFs, which in turn activate RhoA and modulate cytoskeletal organization and cellular motility.

GPCRs are involved in various physiological processes, including neurotransmission, hormone signaling, immune response, and sensory perception. Dysregulation of GPCR function has been implicated in numerous diseases, making them attractive targets for drug development.

"Xenopus" is not a medical term, but it is a genus of highly invasive aquatic frogs native to sub-Saharan Africa. They are often used in scientific research, particularly in developmental biology and genetics. The most commonly studied species is Xenopus laevis, also known as the African clawed frog.

In a medical context, Xenopus might be mentioned when discussing their use in research or as a model organism to study various biological processes or diseases.

Transcription factors are proteins that play a crucial role in regulating gene expression by controlling the transcription of DNA to messenger RNA (mRNA). They function by binding to specific DNA sequences, known as response elements, located in the promoter region or enhancer regions of target genes. This binding can either activate or repress the initiation of transcription, depending on the properties and interactions of the particular transcription factor. Transcription factors often act as part of a complex network of regulatory proteins that determine the precise spatiotemporal patterns of gene expression during development, differentiation, and homeostasis in an organism.

Adaptor proteins are a type of protein that play a crucial role in intracellular signaling pathways by serving as a link between different components of the signaling complex. Specifically, "signal transducing adaptor proteins" refer to those adaptor proteins that are involved in signal transduction processes, where they help to transmit signals from the cell surface receptors to various intracellular effectors. These proteins typically contain modular domains that allow them to interact with multiple partners, thereby facilitating the formation of large signaling complexes and enabling the integration of signals from different pathways.

Signal transducing adaptor proteins can be classified into several families based on their structural features, including the Src homology 2 (SH2) domain, the Src homology 3 (SH3) domain, and the phosphotyrosine-binding (PTB) domain. These domains enable the adaptor proteins to recognize and bind to specific motifs on other signaling molecules, such as receptor tyrosine kinases, G protein-coupled receptors, and cytokine receptors.

One well-known example of a signal transducing adaptor protein is the growth factor receptor-bound protein 2 (Grb2), which contains an SH2 domain that binds to phosphotyrosine residues on activated receptor tyrosine kinases. Grb2 also contains an SH3 domain that interacts with proline-rich motifs on other signaling proteins, such as the guanine nucleotide exchange factor SOS. This interaction facilitates the activation of the Ras small GTPase and downstream signaling pathways involved in cell growth, differentiation, and survival.

Overall, signal transducing adaptor proteins play a critical role in regulating various cellular processes by modulating intracellular signaling pathways in response to extracellular stimuli. Dysregulation of these proteins has been implicated in various diseases, including cancer and inflammatory disorders.

Gastrulation is a fundamental process in embryonic development, characterized by the transformation of a initially flat layer of cells called the blastula into a three-layered structure known as the gastrula. This complex series of cellular movements and rearrangements establishes the foundation for the formation of the three primary germ layers: the ectoderm, mesoderm, and endoderm. These germ layers further differentiate to give rise to all the diverse cell types and tissues in the developing organism, including the nervous system, muscles, bones, and internal organs.

The precise mechanisms of gastrulation vary among different animal groups; however, common features include:

1. Formation of a blastopore: A small indentation or opening that forms on the surface of the blastula, which eventually develops into the primitive gut or anus in the gastrula.
2. Invagination: The process by which cells at the blastopore fold inward and migrate towards the interior of the embryo, forming the endodermal layer.
3. Epiboly: A coordinated movement of cells that spreads over and encloses the yolk within the embryo, contributing to the formation of the ectodermal layer.
4. Delamination: The separation and migration of cells from the epiblast (the outer layer of the blastula) to form the mesodermal layer in between the ectoderm and endoderm.

Gastrulation is a critical period in embryonic development, as errors during this process can lead to severe congenital abnormalities or even embryonic lethality. A thorough understanding of gastrulation has important implications for regenerative medicine, stem cell research, and the study of evolutionary developmental biology (Evo-Devo).

Adenomatous polyposis coli (APC) protein is a tumor suppressor protein that plays a crucial role in regulating cell growth and division. It is encoded by the APC gene, which is located on chromosome 5. The APC protein helps to prevent excessive cell growth and division by inhibiting the activity of a protein called beta-catenin, which promotes cell growth and division when activated.

In individuals with certain genetic disorders, such as familial adenomatous polyposis (FAP), mutations in the APC gene can lead to the production of a defective APC protein or no APC protein at all. This can result in uncontrolled cell growth and division, leading to the development of numerous benign tumors called polyps in the colon and rectum. Over time, some of these polyps may become cancerous, leading to colorectal cancer if left untreated.

APC protein also has other functions in the body, including regulating cell migration and adhesion, and playing a role in maintaining the stability of the cytoskeleton. Mutations in the APC gene have been linked to other types of cancer besides colorectal cancer, including breast, lung, and ovarian cancers.

Morphogenesis is a term used in developmental biology and refers to the process by which cells give rise to tissues and organs with specific shapes, structures, and patterns during embryonic development. This process involves complex interactions between genes, cells, and the extracellular environment that result in the coordinated movement and differentiation of cells into specialized functional units.

Morphogenesis is a dynamic and highly regulated process that involves several mechanisms, including cell proliferation, death, migration, adhesion, and differentiation. These processes are controlled by genetic programs and signaling pathways that respond to environmental cues and regulate the behavior of individual cells within a developing tissue or organ.

The study of morphogenesis is important for understanding how complex biological structures form during development and how these processes can go awry in disease states such as cancer, birth defects, and degenerative disorders.

Bone Morphogenetic Proteins (BMPs) are a group of growth factors that play crucial roles in the development, growth, and repair of bones and other tissues. They belong to the Transforming Growth Factor-β (TGF-β) superfamily and were first discovered when researchers found that certain proteins extracted from demineralized bone matrix had the ability to induce new bone formation.

BMPs stimulate the differentiation of mesenchymal stem cells into osteoblasts, which are the cells responsible for bone formation. They also promote the recruitment and proliferation of these cells, enhancing the overall process of bone regeneration. In addition to their role in bone biology, BMPs have been implicated in various other biological processes, including embryonic development, wound healing, and the regulation of fat metabolism.

There are several types of BMPs (BMP-2, BMP-4, BMP-7, etc.) that exhibit distinct functions and expression patterns. Due to their ability to stimulate bone formation, recombinant human BMPs have been used in clinical applications, such as spinal fusion surgery and non-healing fracture treatment. However, the use of BMPs in medicine has been associated with certain risks and complications, including uncontrolled bone growth, inflammation, and cancer development, which necessitates further research to optimize their therapeutic potential.

Homeodomain proteins are a group of transcription factors that play crucial roles in the development and differentiation of cells in animals and plants. They are characterized by the presence of a highly conserved DNA-binding domain called the homeodomain, which is typically about 60 amino acids long. The homeodomain consists of three helices, with the third helix responsible for recognizing and binding to specific DNA sequences.

Homeodomain proteins are involved in regulating gene expression during embryonic development, tissue maintenance, and organismal growth. They can act as activators or repressors of transcription, depending on the context and the presence of cofactors. Mutations in homeodomain proteins have been associated with various human diseases, including cancer, congenital abnormalities, and neurological disorders.

Some examples of homeodomain proteins include PAX6, which is essential for eye development, HOX genes, which are involved in body patterning, and NANOG, which plays a role in maintaining pluripotency in stem cells.

Transgenic mice are genetically modified rodents that have incorporated foreign DNA (exogenous DNA) into their own genome. This is typically done through the use of recombinant DNA technology, where a specific gene or genetic sequence of interest is isolated and then introduced into the mouse embryo. The resulting transgenic mice can then express the protein encoded by the foreign gene, allowing researchers to study its function in a living organism.

The process of creating transgenic mice usually involves microinjecting the exogenous DNA into the pronucleus of a fertilized egg, which is then implanted into a surrogate mother. The offspring that result from this procedure are screened for the presence of the foreign DNA, and those that carry the desired genetic modification are used to establish a transgenic mouse line.

Transgenic mice have been widely used in biomedical research to model human diseases, study gene function, and test new therapies. They provide a valuable tool for understanding complex biological processes and developing new treatments for a variety of medical conditions.

A cell line that is derived from tumor cells and has been adapted to grow in culture. These cell lines are often used in research to study the characteristics of cancer cells, including their growth patterns, genetic changes, and responses to various treatments. They can be established from many different types of tumors, such as carcinomas, sarcomas, and leukemias. Once established, these cell lines can be grown and maintained indefinitely in the laboratory, allowing researchers to conduct experiments and studies that would not be feasible using primary tumor cells. It is important to note that tumor cell lines may not always accurately represent the behavior of the original tumor, as they can undergo genetic changes during their time in culture.

According to the National Institutes of Health (NIH), stem cells are "initial cells" or "precursor cells" that have the ability to differentiate into many different cell types in the body. They can also divide without limit to replenish other cells for as long as the person or animal is still alive.

There are two main types of stem cells: embryonic stem cells, which come from human embryos, and adult stem cells, which are found in various tissues throughout the body. Embryonic stem cells have the ability to differentiate into all cell types in the body, while adult stem cells have more limited differentiation potential.

Stem cells play an essential role in the development and repair of various tissues and organs in the body. They are currently being studied for their potential use in the treatment of a wide range of diseases and conditions, including cancer, diabetes, heart disease, and neurological disorders. However, more research is needed to fully understand the properties and capabilities of these cells before they can be used safely and effectively in clinical settings.

Lithium Chloride (LiCl) is not typically defined in a medical context as it is not a medication or a clinical condition. However, it can be defined chemically as an inorganic compound consisting of lithium and chlorine. Its chemical formula is LiCl, and it is commonly used in laboratory settings for various purposes such as a drying agent or a component in certain chemical reactions.

It's worth noting that while lithium salts like lithium carbonate (Li2CO3) are used medically to treat bipolar disorder, lithium chloride is not used for this purpose due to its higher toxicity compared to other lithium salts.

Hedgehog proteins are a group of signaling molecules that play crucial roles in the development and regulation of various biological processes in animals. They are named after the hedgehog mutant fruit flies, which have spiky bristles due to defects in this pathway. These proteins are involved in cell growth, differentiation, and tissue regeneration. They exert their effects by binding to specific receptors on the surface of target cells, leading to a cascade of intracellular signaling events that ultimately influence gene expression and cell behavior.

There are three main types of Hedgehog proteins in mammals: Sonic hedgehog (Shh), Indian hedgehog (Ihh), and Desert hedgehog (Dhh). These protecules undergo post-translational modifications, including cleavage and lipid modification, which are essential for their activity. Dysregulation of Hedgehog signaling has been implicated in various diseases, including cancer, developmental abnormalities, and degenerative disorders.

'Cell lineage' is a term used in biology and medicine to describe the developmental history or relationship of a cell or group of cells to other cells, tracing back to the original progenitor or stem cell. It refers to the series of cell divisions and differentiation events that give rise to specific types of cells in an organism over time.

In simpler terms, cell lineage is like a family tree for cells, showing how they are related to each other through a chain of cell division and specialization events. This concept is important in understanding the development, growth, and maintenance of tissues and organs in living beings.

HEK293 cells, also known as human embryonic kidney 293 cells, are a line of cells used in scientific research. They were originally derived from human embryonic kidney cells and have been adapted to grow in a lab setting. HEK293 cells are widely used in molecular biology and biochemistry because they can be easily transfected (a process by which DNA is introduced into cells) and highly express foreign genes. As a result, they are often used to produce proteins for structural and functional studies. It's important to note that while HEK293 cells are derived from human tissue, they have been grown in the lab for many generations and do not retain the characteristics of the original embryonic kidney cells.

A cell line is a culture of cells that are grown in a laboratory for use in research. These cells are usually taken from a single cell or group of cells, and they are able to divide and grow continuously in the lab. Cell lines can come from many different sources, including animals, plants, and humans. They are often used in scientific research to study cellular processes, disease mechanisms, and to test new drugs or treatments. Some common types of human cell lines include HeLa cells (which come from a cancer patient named Henrietta Lacks), HEK293 cells (which come from embryonic kidney cells), and HUVEC cells (which come from umbilical vein endothelial cells). It is important to note that cell lines are not the same as primary cells, which are cells that are taken directly from a living organism and have not been grown in the lab.

Repressor proteins are a type of regulatory protein in molecular biology that suppress the transcription of specific genes into messenger RNA (mRNA) by binding to DNA. They function as part of gene regulation processes, often working in conjunction with an operator region and a promoter region within the DNA molecule. Repressor proteins can be activated or deactivated by various signals, allowing for precise control over gene expression in response to changing cellular conditions.

There are two main types of repressor proteins:

1. DNA-binding repressors: These directly bind to specific DNA sequences (operator regions) near the target gene and prevent RNA polymerase from transcribing the gene into mRNA.
2. Allosteric repressors: These bind to effector molecules, which then cause a conformational change in the repressor protein, enabling it to bind to DNA and inhibit transcription.

Repressor proteins play crucial roles in various biological processes, such as development, metabolism, and stress response, by controlling gene expression patterns in cells.

Biological models, also known as physiological models or organismal models, are simplified representations of biological systems, processes, or mechanisms that are used to understand and explain the underlying principles and relationships. These models can be theoretical (conceptual or mathematical) or physical (such as anatomical models, cell cultures, or animal models). They are widely used in biomedical research to study various phenomena, including disease pathophysiology, drug action, and therapeutic interventions.

Examples of biological models include:

1. Mathematical models: These use mathematical equations and formulas to describe complex biological systems or processes, such as population dynamics, metabolic pathways, or gene regulation networks. They can help predict the behavior of these systems under different conditions and test hypotheses about their underlying mechanisms.
2. Cell cultures: These are collections of cells grown in a controlled environment, typically in a laboratory dish or flask. They can be used to study cellular processes, such as signal transduction, gene expression, or metabolism, and to test the effects of drugs or other treatments on these processes.
3. Animal models: These are living organisms, usually vertebrates like mice, rats, or non-human primates, that are used to study various aspects of human biology and disease. They can provide valuable insights into the pathophysiology of diseases, the mechanisms of drug action, and the safety and efficacy of new therapies.
4. Anatomical models: These are physical representations of biological structures or systems, such as plastic models of organs or tissues, that can be used for educational purposes or to plan surgical procedures. They can also serve as a basis for developing more sophisticated models, such as computer simulations or 3D-printed replicas.

Overall, biological models play a crucial role in advancing our understanding of biology and medicine, helping to identify new targets for therapeutic intervention, develop novel drugs and treatments, and improve human health.

A mutation is a permanent change in the DNA sequence of an organism's genome. Mutations can occur spontaneously or be caused by environmental factors such as exposure to radiation, chemicals, or viruses. They may have various effects on the organism, ranging from benign to harmful, depending on where they occur and whether they alter the function of essential proteins. In some cases, mutations can increase an individual's susceptibility to certain diseases or disorders, while in others, they may confer a survival advantage. Mutations are the driving force behind evolution, as they introduce new genetic variability into populations, which can then be acted upon by natural selection.

Reverse Transcriptase Polymerase Chain Reaction (RT-PCR) is a laboratory technique used in molecular biology to amplify and detect specific DNA sequences. This technique is particularly useful for the detection and quantification of RNA viruses, as well as for the analysis of gene expression.

The process involves two main steps: reverse transcription and polymerase chain reaction (PCR). In the first step, reverse transcriptase enzyme is used to convert RNA into complementary DNA (cDNA) by reading the template provided by the RNA molecule. This cDNA then serves as a template for the PCR amplification step.

In the second step, the PCR reaction uses two primers that flank the target DNA sequence and a thermostable polymerase enzyme to repeatedly copy the targeted cDNA sequence. The reaction mixture is heated and cooled in cycles, allowing the primers to anneal to the template, and the polymerase to extend the new strand. This results in exponential amplification of the target DNA sequence, making it possible to detect even small amounts of RNA or cDNA.

RT-PCR is a sensitive and specific technique that has many applications in medical research and diagnostics, including the detection of viruses such as HIV, hepatitis C virus, and SARS-CoV-2 (the virus that causes COVID-19). It can also be used to study gene expression, identify genetic mutations, and diagnose genetic disorders.

A gastrula is a stage in the early development of many animals, including humans, that occurs following fertilization and cleavage of the zygote. During this stage, the embryo undergoes a process called gastrulation, which involves a series of cell movements that reorganize the embryo into three distinct layers: the ectoderm, mesoderm, and endoderm. These germ layers give rise to all the different tissues and organs in the developing organism.

The gastrula is characterized by the presence of a central cavity called the archenteron, which will eventually become the gut or gastrointestinal tract. The opening of the archenteron is called the blastopore, which will give rise to either the mouth or anus, depending on the animal group.

In summary, a gastrula is a developmental stage in which an embryo undergoes gastrulation to form three germ layers and a central cavity, which will eventually develop into various organs and tissues of the body.

Proteins are complex, large molecules that play critical roles in the body's functions. They are made up of amino acids, which are organic compounds that are the building blocks of proteins. Proteins are required for the structure, function, and regulation of the body's tissues and organs. They are essential for the growth, repair, and maintenance of body tissues, and they play a crucial role in many biological processes, including metabolism, immune response, and cellular signaling. Proteins can be classified into different types based on their structure and function, such as enzymes, hormones, antibodies, and structural proteins. They are found in various foods, especially animal-derived products like meat, dairy, and eggs, as well as plant-based sources like beans, nuts, and grains.

A mammalian embryo is the developing offspring of a mammal, from the time of implantation of the fertilized egg (blastocyst) in the uterus until the end of the eighth week of gestation. During this period, the embryo undergoes rapid cell division and organ differentiation to form a complex structure with all the major organs and systems in place. This stage is followed by fetal development, which continues until birth. The study of mammalian embryos is important for understanding human development, evolution, and reproductive biology.

Cell polarity refers to the asymmetric distribution of membrane components, cytoskeleton, and organelles in a cell. This asymmetry is crucial for various cellular functions such as directed transport, cell division, and signal transduction. The plasma membrane of polarized cells exhibits distinct domains with unique protein and lipid compositions that define apical, basal, and lateral surfaces of the cell.

In epithelial cells, for example, the apical surface faces the lumen or external environment, while the basolateral surface interacts with other cells or the extracellular matrix. The establishment and maintenance of cell polarity are regulated by various factors including protein complexes, lipids, and small GTPases. Loss of cell polarity has been implicated in several diseases, including cancer and neurological disorders.

Phosphoproteins are proteins that have been post-translationally modified by the addition of a phosphate group (-PO3H2) onto specific amino acid residues, most commonly serine, threonine, or tyrosine. This process is known as phosphorylation and is mediated by enzymes called kinases. Phosphoproteins play crucial roles in various cellular processes such as signal transduction, cell cycle regulation, metabolism, and gene expression. The addition or removal of a phosphate group can activate or inhibit the function of a protein, thereby serving as a switch to control its activity. Phosphoproteins can be detected and quantified using techniques such as Western blotting, mass spectrometry, and immunofluorescence.

Glycoproteins are complex proteins that contain oligosaccharide chains (glycans) covalently attached to their polypeptide backbone. These glycans are linked to the protein through asparagine residues (N-linked) or serine/threonine residues (O-linked). Glycoproteins play crucial roles in various biological processes, including cell recognition, cell-cell interactions, cell adhesion, and signal transduction. They are widely distributed in nature and can be found on the outer surface of cell membranes, in extracellular fluids, and as components of the extracellular matrix. The structure and composition of glycoproteins can vary significantly depending on their function and location within an organism.

Embryonic induction is a process that occurs during the development of a multicellular organism, where one group of cells in the embryo signals and influences the developmental fate of another group of cells. This interaction leads to the formation of specific structures or organs in the developing embryo. The signaling cells that initiate the process are called organizers, and they release signaling molecules known as morphogens that bind to receptors on the target cells and trigger a cascade of intracellular signals that ultimately lead to changes in gene expression and cell fate. Embryonic induction is a crucial step in the development of complex organisms and plays a key role in establishing the body plan and organizing the different tissues and organs in the developing embryo.

Ectoderm is the outermost of the three primary germ layers in a developing embryo, along with the endoderm and mesoderm. The ectoderm gives rise to the outer covering of the body, including the skin, hair, nails, glands, and the nervous system, which includes the brain, spinal cord, and peripheral nerves. It also forms the lining of the mouth, anus, nose, and ears. Essentially, the ectoderm is responsible for producing all the epidermal structures and the neural crest cells that contribute to various derivatives such as melanocytes, adrenal medulla, smooth muscle, and peripheral nervous system components.

'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.

Neoplastic gene expression regulation refers to the processes that control the production of proteins and other molecules from genes in neoplastic cells, or cells that are part of a tumor or cancer. In a normal cell, gene expression is tightly regulated to ensure that the right genes are turned on or off at the right time. However, in cancer cells, this regulation can be disrupted, leading to the overexpression or underexpression of certain genes.

Neoplastic gene expression regulation can be affected by a variety of factors, including genetic mutations, epigenetic changes, and signals from the tumor microenvironment. These changes can lead to the activation of oncogenes (genes that promote cancer growth and development) or the inactivation of tumor suppressor genes (genes that prevent cancer).

Understanding neoplastic gene expression regulation is important for developing new therapies for cancer, as targeting specific genes or pathways involved in this process can help to inhibit cancer growth and progression.

"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.

Embryonic development is the series of growth and developmental stages that occur during the formation and early growth of the embryo. In humans, this stage begins at fertilization (when the sperm and egg cell combine) and continues until the end of the 8th week of pregnancy. During this time, the fertilized egg (now called a zygote) divides and forms a blastocyst, which then implants into the uterus. The cells in the blastocyst begin to differentiate and form the three germ layers: the ectoderm, mesoderm, and endoderm. These germ layers will eventually give rise to all of the different tissues and organs in the body.

Embryonic development is a complex and highly regulated process that involves the coordinated interaction of genetic and environmental factors. It is characterized by rapid cell division, migration, and differentiation, as well as programmed cell death (apoptosis) and tissue remodeling. Abnormalities in embryonic development can lead to birth defects or other developmental disorders.

It's important to note that the term "embryo" is used to describe the developing organism from fertilization until the end of the 8th week of pregnancy in humans, after which it is called a fetus.

Osteoblasts are specialized bone-forming cells that are derived from mesenchymal stem cells. They play a crucial role in the process of bone formation and remodeling. Osteoblasts synthesize, secrete, and mineralize the organic matrix of bones, which is mainly composed of type I collagen.

These cells have receptors for various hormones and growth factors that regulate their activity, such as parathyroid hormone, vitamin D, and transforming growth factor-beta. When osteoblasts are not actively producing bone matrix, they can become trapped within the matrix they produce, where they differentiate into osteocytes, which are mature bone cells that play a role in maintaining bone structure and responding to mechanical stress.

Abnormalities in osteoblast function can lead to various bone diseases, such as osteoporosis, osteogenesis imperfecta, and Paget's disease of bone.

Genetically modified animals (GMAs) are those whose genetic makeup has been altered using biotechnological techniques. This is typically done by introducing one or more genes from another species into the animal's genome, resulting in a new trait or characteristic that does not naturally occur in that species. The introduced gene is often referred to as a transgene.

The process of creating GMAs involves several steps:

1. Isolation: The desired gene is isolated from the DNA of another organism.
2. Transfer: The isolated gene is transferred into the target animal's cells, usually using a vector such as a virus or bacterium.
3. Integration: The transgene integrates into the animal's chromosome, becoming a permanent part of its genetic makeup.
4. Selection: The modified cells are allowed to multiply, and those that contain the transgene are selected for further growth and development.
5. Breeding: The genetically modified individuals are bred to produce offspring that carry the desired trait.

GMAs have various applications in research, agriculture, and medicine. In research, they can serve as models for studying human diseases or testing new therapies. In agriculture, GMAs can be developed to exhibit enhanced growth rates, improved disease resistance, or increased nutritional value. In medicine, GMAs may be used to produce pharmaceuticals or other therapeutic agents within their bodies.

Examples of genetically modified animals include mice with added genes for specific proteins that make them useful models for studying human diseases, goats that produce a human protein in their milk to treat hemophilia, and pigs with enhanced resistance to certain viruses that could potentially be used as organ donors for humans.

It is important to note that the use of genetically modified animals raises ethical concerns related to animal welfare, environmental impact, and potential risks to human health. These issues must be carefully considered and addressed when developing and implementing GMA technologies.

Gene knockdown techniques are methods used to reduce the expression or function of specific genes in order to study their role in biological processes. These techniques typically involve the use of small RNA molecules, such as siRNAs (small interfering RNAs) or shRNAs (short hairpin RNAs), which bind to and promote the degradation of complementary mRNA transcripts. This results in a decrease in the production of the protein encoded by the targeted gene.

Gene knockdown techniques are often used as an alternative to traditional gene knockout methods, which involve completely removing or disrupting the function of a gene. Knockdown techniques allow for more subtle and reversible manipulation of gene expression, making them useful for studying genes that are essential for cell survival or have redundant functions.

These techniques are widely used in molecular biology research to investigate gene function, genetic interactions, and disease mechanisms. However, it is important to note that gene knockdown can have off-target effects and may not completely eliminate the expression of the targeted gene, so results should be interpreted with caution.

No FAQ available that match "wnt3 protein"

No images available that match "wnt3 protein"