Cyclin D1
Cyclin D2
Cyclin D3
Cyclin A
Cyclin D
Cyclin E
Cyclin B
Cyclin B1
Cyclin A1
Cyclins
Cyclin A2
Cyclin-Dependent Kinase 4
Cyclin-Dependent Kinases
Cyclin G1
Cyclin G
Cell Cycle
Cyclin-Dependent Kinase 2
G1 Phase
Genes, bcl-1
Cyclin C
Retinoblastoma Protein
CDC2-CDC28 Kinases
Cyclin B2
Cyclin T
Oncogene Proteins
Cyclin H
Cell Cycle Proteins
S Phase
Cyclin G2
CDC2 Protein Kinase
Cyclin-Dependent Kinase Inhibitor p21
Phosphorylation
Cell Division
Protein-Serine-Threonine Kinases
Cyclin-Dependent Kinase Inhibitor p27
Proto-Oncogene Proteins
Gene Expression Regulation, Neoplastic
Tumor Suppressor Proteins
Cyclin I
RNA, Messenger
Mitosis
Immunohistochemistry
Tumor Cells, Cultured
Lymphoma, Mantle-Cell
Signal Transduction
Down-Regulation
Cyclin-Dependent Kinase Inhibitor p16
Blotting, Western
Promoter Regions, Genetic
Transfection
Proliferating Cell Nuclear Antigen
Cell Nucleus
Apoptosis
Gene Expression Regulation
G2 Phase
E2F Transcription Factors
beta Catenin
Cyclin-Dependent Kinase 6
Transcription Factors
3T3 Cells
Protein Binding
Glycogen Synthase Kinase 3
Ki-67 Antigen
Cell Transformation, Neoplastic
Transcription, Genetic
F-Box Proteins
Cells, Cultured
Tumor Suppressor Protein p53
Reverse Transcriptase Polymerase Chain Reaction
Molecular Sequence Data
DNA-Binding Proteins
RNA, Small Interfering
Proto-Oncogene Proteins c-myc
NIH 3T3 Cells
E2F1 Transcription Factor
Mutation
Nuclear Proteins
Enzyme Activation
Enzyme Inhibitors
Base Sequence
Flow Cytometry
Transcription Factor DP1
Fibroblasts
Chromosomes, Human, Pair 11
Gene Expression
Up-Regulation
Retinoblastoma-Like Protein p107
cdc25 Phosphatases
Neoplasm Proteins
Retinoblastoma-Binding Protein 1
Proteasome Endopeptidase Complex
S-Phase Kinase-Associated Proteins
HeLa Cells
Cell Cycle Checkpoints
Tumor Markers, Biological
Mice, Transgenic
Trans-Activators
Recombinant Fusion Proteins
Immunoblotting
Proto-Oncogene Proteins c-akt
Precipitin Tests
DNA Primers
Protein Kinases
Amino Acid Sequence
RNA Interference
Cell Differentiation
Epithelial Cells
Transcriptional Activation
Microtubule-Associated Proteins
Chromosomes, Human, Pair 14
Models, Biological
Protamine Kinase
Carrier Proteins
Cyclin-Dependent Kinase Inhibitor Proteins
Threonine
Carcinoma, Squamous Cell
G1 Phase Cell Cycle Checkpoints
Phosphatidylinositol 3-Kinases
Maturation-Promoting Factor
Mitogen-Activated Protein Kinases
Mice, Knockout
Luciferases
DNA
Genes, myc
Immunoprecipitation
Mice, Nude
Active Transport, Cell Nucleus
Cyclin-Dependent Kinase 9
Bromodeoxyuridine
Ubiquitins
Cytoplasm
Gene Amplification
E2F4 Transcription Factor
Ubiquitin-Protein Ligase Complexes
Blotting, Northern
Prognosis
Coupling of the cell cycle and myogenesis through the cyclin D1-dependent interaction of MyoD with cdk4. (1/3253)
Proliferating myoblasts express the muscle determination factor, MyoD, throughout the cell cycle in the absence of differentiation. Here we show that a mitogen-sensitive mechanism, involving the direct interaction between MyoD and cdk4, restricts myoblast differentiation to cells that have entered into the G0 phase of the cell cycle under mitogen withdrawal. Interaction between MyoD and cdk4 disrupts MyoD DNA-binding, muscle-specific gene activation and myogenic conversion of 10T1/2 cells independently of cyclin D1 and the CAK activation of cdk4. Forced induction of cyclin D1 in myotubes results in the cytoplasmic to nuclear translocation of cdk4. The specific MyoD-cdk4 interaction in dividing myoblasts, coupled with the cyclin D1-dependent nuclear targeting of cdk4, suggests a mitogen-sensitive mechanism whereby cyclin D1 can regulate MyoD function and the onset of myogenesis by controlling the cellular location of cdk4 rather than the phosphorylation status of MyoD. (+info)Differential roles for cyclin-dependent kinase inhibitors p21 and p16 in the mechanisms of senescence and differentiation in human fibroblasts. (2/3253)
The irreversible G1 arrest in senescent human diploid fibroblasts is probably caused by inactivation of the G1 cyclin-cyclin-dependent kinase (Cdk) complexes responsible for phosphorylation of the retinoblastoma protein (pRb). We show that the Cdk inhibitor p21(Sdi1,Cip1,Waf1), which accumulates progressively in aging cells, binds to and inactivates all cyclin E-Cdk2 complexes in senescent cells, whereas in young cells only p21-free Cdk2 complexes are active. Furthermore, the senescent-cell-cycle arrest occurs prior to the accumulation of the Cdk4-Cdk6 inhibitor p16(Ink4a), suggesting that p21 may be sufficient for this event. Accordingly, cyclin D1-associated phosphorylation of pRb at Ser-780 is lacking even in newly senescent fibroblasts that have a low amount of p16. Instead, the cyclin D1-Cdk4 and cyclin D1-Cdk6 complexes in these cells are associated with an increased amount of p21, suggesting that p21 may be responsible for inactivation of both cyclin E- and cyclin D1-associated kinase activity at the early stage of senescence. Moreover, even in the late stage of senescence when p16 is high, cyclin D1-Cdk4 complexes are persistent, albeit reduced by +info)Progesterone inhibits estrogen-induced cyclin D1 and cdk4 nuclear translocation, cyclin E- and cyclin A-cdk2 kinase activation, and cell proliferation in uterine epithelial cells in mice. (3/3253)
The response of the uterine epithelium to female sex steroid hormones provides an excellent model to study cell proliferation in vivo since both stimulation and inhibition of cell proliferation can be studied. Thus, when administered to ovariectomized adult mice 17beta-estradiol (E2) stimulates a synchronized wave of DNA synthesis and cell division in the epithelial cells, while pretreatment with progesterone (P4) completely inhibits this E2-induced cell proliferation. Using a simple method to isolate the uterine epithelium with high purity, we have shown that E2 treatment induces a relocalization of cyclin D1 and, to a lesser extent, cdk4 from the cytoplasm into the nucleus and results in the orderly activation of cyclin E- and cyclin A-cdk2 kinases and hyperphosphorylation of pRb and p107. P4 pretreatment did not alter overall levels of cyclin D1, cdk4, or cdk6 nor their associated kinase activities but instead inhibited the E2-induced nuclear localization of cyclin D1 to below the control level and, to a lesser extent, nuclear cdk4 levels, with a consequent inhibition of pRb and p107 phosphorylation. In addition, it abrogated E2-induced cyclin E-cdk2 activation by dephosphorylation of cdk2, followed by inhibition of cyclin A expression and consequently of cyclin A-cdk2 kinase activity and further inhibition of phosphorylation of pRb and p107. P4 is used therapeutically to oppose the effect of E2 during hormone replacement therapy and in the treatment of uterine adenocarcinoma. This study showing a novel mechanism of cell cycle inhibition by P4 may provide the basis for the development of new antiestrogens. (+info)Insertion of excised IgH switch sequences causes overexpression of cyclin D1 in a myeloma tumor cell. (4/3253)
Oncogenes are often dysregulated in B cell tumors as a result of a reciprocal translocation involving an immunoglobulin locus. The translocations are caused by errors in two developmentally regulated DNA recombination processes: V(D)J and IgH switch recombination. Both processes share the property of joining discontinuous sequences from one chromosome and releasing intervening sequences as circles that are lost from progeny cells. Here we show that these intervening sequences may instead insert in the genome and that during productive IgH mu-epsilon switch recombination in U266 myeloma tumor cells, a portion of the excised IgH switch intervening sequences containing the 3' alpha-1 enhancer has inserted on chromosome 11q13, resulting in overexpression of the adjacent cyclin D1 oncogene. (+info)Alternatively spliced EDA segment regulates fibronectin-dependent cell cycle progression and mitogenic signal transduction. (5/3253)
Fibronectin (FN) is comprised of multiple isoforms arising from alternative splicing of a single gene transcript. One of the alternatively spliced segments, EDA, is expressed prominently in embryonic development, malignant transformation, and wound healing. We showed previously that EDA+ FN was more potent than EDA- FN in promoting cell spreading and cell migration because of its enhanced binding affinity to integrin alpha5beta1 (Manabe, R., Oh-e, N., Maeda, T., Fukuda, T., and Sekiguchi, K. (1997) J. Cell Biol. 139, 295-307). In this study, we compared the cell cycle progression and its associated signal transduction events induced by FN isoforms with or without the EDA segment to examine whether the EDA segment modulates the cell proliferative potential of FN. We found that EDA+ FN was more potent than EDA- FN in inducing G1-S phase transition. Inclusion of the EDA segment potentiated the ability of FN to induce expression of cyclin D1, hyperphosphorylation of pRb, and activation of mitogen-activated protein kinase extracellular signal regulated kinase 2 (ERK2). EDA+ FN was also more potent than EDA- FN in promoting FN-mediated tyrosine phosphorylation of p130(Cas), but not focal adhesion kinase, which occurred in parallel with the activation of ERK2, suggesting that p130(Cas) may be involved in activation of ERK2. These results indicated that alternative splicing at the EDA region is a novel mechanism that promotes FN-induced cell cycle progression through up-regulation of integrin-mediated mitogenic signal transduction. (+info)Survey of gene amplifications during prostate cancer progression by high-throughout fluorescence in situ hybridization on tissue microarrays. (6/3253)
Prostate cancer development and progression is driven by the accumulation of genetic changes, the nature of which remains incompletely understood To facilitate high-throughput analysis of molecular events taking place in primary, recurrent, and metastat prostate cancer, we constructed a tissue microarray containing small 0.6-mm cylindrical samples acquired from 371 formalin-fixed blocks, including benign prostatic hyperplasia (n = 32) and primary tumors (n = 223), as well as both locally recurrent tumors (n = 54) and metastases (n = 62) from patients with hormone-refractory disease. Fluorescence in situ hybridization (FISH) was applied to the analysis of consecutive tissue microarray sections with probes for five different genes. High-level (> or =3X) amplifications were very rare (<2%) in primary prostate cancers However, in metastases from patients with hormone-refractory disease, amplification of the androgen receptor gene was seen in 22%, MYC in 11%, and Cyclin-D1 in 5% of the cases. In specimens from locally recurrent tumors, the corresponding percentages were 23, 4, and 8%. ERBB2 and NMYC amplifications were never detected at any stage of prostate cancer progression. In conclusion, FISH to tissue microarray sections enables high-throughput analysis of genetic alterations contributing to cancer development and progression. Our results implicate a role for amplification of androgen receptor in hormonal therapy failure and that of MYC in the metastatic progression of human prostate cancer. (+info)Alterations of Rb pathway (Rb-p16INK4-cyclin D1) in preinvasive bronchial lesions. (7/3253)
Lung cancer results from a stepwise accumulation of genetic and molecular abnormalities with unknown temporal relationships to precursor bronchial lesions. In a search for biomarkers of malignant progression, we analyzed the expression of the tumor suppressor gene Rb and of the proteins regulating its phosphorylation and function in G1 arrest, p16INK4A and cyclin D1, in preinvasive bronchial lesions accompanying cancer in 75 patients, in comparison with similar lesions in 22 patients with no cancer history. Rb was constantly expressed in preinvasive lesions, including carcinoma in situ (CIS). In contrast, p16 expression was lost in moderate dysplasia (12%) and in CIS (30%) in patients with lung cancer. p16 loss occurred exclusively in patients who displayed loss of p16 expression in their related invasive carcinoma. Loss of p16 expression was not seen in nine patients with dysplasia but no cancer progression. Cyclin D1 overexpression was seen in hyperplasia and metaplasia (6%), mild dysplasia (17%), moderate dysplasia (46%), and CIS (38%) in patients with cancer but was lost in 5% of the patients during the process of invasion; it was also observed in patients with no cancer progression (14%). Our results indicate that Rb protein function can be invalidated before invasion through alteration of the Rb phosphorylation pathway, by p16 inhibition, and/or by cyclin D1 overexpression and suggest a role for p16 and cyclin D1 deregulation in progression of preinvasive bronchial lesions to invasive carcinoma. (+info)Cyclin D1 proteolysis: a retinoid chemoprevention signal in normal, immortalized, and transformed human bronchial epithelial cells. (8/3253)
BACKGROUND: Retinoids (derivatives of vitamin A) are reported to reduce the occurrence of some second primary cancers, including aerodigestive tract tumors. In contrast, beta-carotene does not reduce the occurrence of primary aerodigestive tract cancers. Mechanisms explaining these effective retinoid and ineffective carotenoid chemoprevention results are poorly defined. Recently, the all-trans-retinoic acid (RA)-induced proteolysis of cyclin D1 that leads to the arrest of cells in G1 phase of the cell cycle was described in human bronchial epithelial cells and is a promising candidate for such a mechanism. In this study, we have investigated this proteolysis as a common signal used by carotenoids or receptor-selective and receptor-nonselective retinoids. METHODS: We treated cultured normal human bronchial epithelial cells, immortalized human bronchial epithelial cells (BEAS-2B), and transformed human bronchial epithelial cells (BEAS-2BNNK) with receptor-selective or receptor-nonselective retinoids or with carotenoids and studied the effects on cell proliferation by means of tritiated thymidine incorporation and on cyclin D1 expression by means of immunoblot analysis. We also examined whether calpain inhibitor I, an inhibitor of the 26S proteasome degradation pathway, affected the decline (i.e., proteolysis) of cyclin D1. RESULTS: Receptor-nonselective retinoids were superior to the carotenoids studied in mediating the decline in cyclin D1 expression and in suppressing the growth of bronchial epithelial cells. Retinoids that activated retinoic acid receptor beta or retinoid X receptor pathways preferentially led to a decrease in the amount of cyclin D1 protein and a corresponding decline in growth. The retinoid-mediated degradation of cyclin D1 was blocked by cotreatment with calpain inhibitor I. CONCLUSIONS: Retinoid-dependent cyclin D1 proteolysis is a common chemoprevention signal in normal and neoplastic human bronchial epithelial cells. In contrast, carotenoids did not affect cyclin D1 expression. Thus, the degradation of cyclin D1 is a candidate intermediate marker for effective retinoid-mediated cancer chemoprevention in the aerodigestive tract. (+info)Cyclin D1 is a protein that plays a critical role in regulating the progression of the cell cycle from the G1 phase to the S phase. It is encoded by the CCND1 gene and is expressed in a variety of tissues, including epithelial cells, fibroblasts, and leukocytes. In the cell cycle, cyclin D1 binds to and activates cyclin-dependent kinases (CDKs), particularly CDK4 and CDK6. This complex then phosphorylates retinoblastoma protein (Rb), which releases the transcription factor E2F from its inhibition. E2F then activates the transcription of genes required for DNA synthesis and cell proliferation. Abnormal expression or activity of cyclin D1 has been implicated in the development of various types of cancer, including breast, prostate, and lung cancer. Overexpression of cyclin D1 can lead to uncontrolled cell proliferation and the formation of tumors. Conversely, loss of cyclin D1 function has been associated with cell cycle arrest and the development of cancer.
Cyclin D2 is a protein that plays a role in regulating the cell cycle, which is the process by which cells grow, divide, and replicate their genetic material. Cyclin D2 is expressed at high levels in cells that are actively dividing, and it helps to promote the progression of the cell cycle from the G1 phase (the first phase of interphase) to the S phase (the second phase of interphase), where DNA replication occurs. In the medical field, cyclin D2 is often studied in the context of cancer. Abnormal expression or activity of cyclin D2 has been linked to the development and progression of various types of cancer, including breast cancer, ovarian cancer, and prostate cancer. In these cases, cyclin D2 may contribute to uncontrolled cell growth and division, leading to the formation of tumors. Cyclin D2 is also being studied as a potential therapeutic target in cancer treatment. Researchers are exploring the use of drugs that inhibit the activity of cyclin D2 as a way to slow or stop the growth of cancer cells.
Cyclin D3 is a protein that plays a role in regulating the progression of the cell cycle, which is the process by which cells grow and divide. It is a type of cyclin, which are proteins that are involved in regulating the cell cycle by interacting with cyclin-dependent kinases (CDKs). Cyclin D3 is expressed primarily in cells that are actively dividing, such as those in the skin, bone marrow, and breast. It helps to promote the progression of the cell cycle from the G1 phase (the first phase of the cell cycle) to the S phase (the second phase), where DNA replication occurs. Abnormal expression of cyclin D3 has been linked to the development of certain types of cancer, including breast, prostate, and colon cancer.
Cyclin A is a protein that plays a crucial role in regulating the cell cycle, which is the process by which cells grow, divide, and replicate their genetic material. Cyclin A is synthesized in the S phase of the cell cycle, when the cell is preparing to divide, and is degraded as the cell enters the G2 phase, before it actually divides. Cyclin A forms a complex with the cyclin-dependent kinase (CDK) 2, which is a key regulator of the cell cycle. This complex phosphorylates a variety of target proteins, including the retinoblastoma protein (Rb), which is a tumor suppressor that prevents cells from dividing unless they have completed the necessary DNA replication and repair processes. When Cyclin A and CDK2 are activated, they promote the progression of the cell cycle from the S phase to the G2 phase, and ultimately to mitosis, the process by which the cell divides into two daughter cells. Dysregulation of Cyclin A expression or activity has been implicated in a variety of diseases, including cancer, where it can contribute to uncontrolled cell proliferation and tumor growth.
Cyclin D is a protein that plays a critical role in regulating the progression of the cell cycle, which is the process by which cells divide and replicate. Cyclin D is synthesized in response to growth signals and helps to promote the transition of cells from the G1 phase (interphase) to the S phase (synthesis phase) of the cell cycle. During the S phase, the cell replicates its DNA in preparation for cell division. Cyclin D is often overexpressed in cancer cells, leading to uncontrolled cell proliferation and the development of tumors. In addition, mutations in the genes that encode cyclin D or its regulatory proteins can also contribute to the development of cancer. Cyclin D is a target for several cancer therapies, including targeted therapies that block the activity of cyclin D or its downstream signaling pathways. Understanding the role of cyclin D in the cell cycle and its role in cancer is an active area of research in the medical field.
Cyclin E is a protein that plays a crucial role in regulating the cell cycle, which is the process by which cells grow, divide, and replicate their genetic material. Cyclin E is synthesized in the late G1 phase of the cell cycle and is degraded as the cell enters the S phase, where DNA replication occurs. Cyclin E functions by binding to and activating the cyclin-dependent kinase (CDK) 2, which is a key regulator of the G1/S transition. The cyclin E-CDK2 complex phosphorylates several target proteins, including the retinoblastoma protein (Rb), which is a tumor suppressor that inhibits cell cycle progression. When cyclin E-CDK2 is activated, it phosphorylates Rb, releasing it from its inhibitory complex and allowing the cell to progress into the S phase. Abnormal expression or activity of cyclin E has been implicated in the development of several types of cancer, including breast, ovarian, and cervical cancer. In these cancers, high levels of cyclin E can lead to uncontrolled cell proliferation and the formation of tumors. Therefore, cyclin E is an important target for cancer therapy, and several drugs that target cyclin E or its downstream targets are currently being developed for the treatment of cancer.
Cyclin B is a protein that plays a crucial role in regulating the progression of the cell cycle, particularly during the M phase (mitosis). It is synthesized and degraded in a tightly regulated manner, with its levels increasing just before the onset of mitosis and decreasing afterwards. Cyclin B forms a complex with the cyclin-dependent kinase (CDK) 1, which is also known as Cdk1. This complex is responsible for phosphorylating various target proteins, including the nuclear envelope, kinetochores, and microtubules, which are essential for the proper progression of mitosis. Disruptions in the regulation of cyclin B and CDK1 activity can lead to various diseases, including cancer. For example, overexpression of cyclin B or mutations in CDK1 can result in uncontrolled cell proliferation and the development of tumors. Conversely, loss of cyclin B function can lead to cell cycle arrest and genomic instability, which can also contribute to cancer development.
Cyclin B1 is a protein that plays a crucial role in regulating the progression of the cell cycle, particularly during the M phase (mitosis). It is synthesized and degraded in a tightly regulated manner, with its levels increasing just before the onset of mitosis and decreasing afterwards. Cyclin B1 forms a complex with the cyclin-dependent kinase (CDK) 1, which is a key regulator of cell division. This complex phosphorylates various target proteins, including the nuclear envelope, microtubules, and other cell cycle regulators, to promote the progression of mitosis. Mutations in the gene encoding cyclin B1 have been implicated in several human diseases, including cancer. In particular, overexpression of cyclin B1 has been observed in many types of cancer, and it has been proposed that this contributes to uncontrolled cell proliferation and tumor growth.
Cyclin A1 is a protein that plays a role in regulating the cell cycle, which is the process by which cells grow, divide, and replicate their genetic material. Cyclin A1 is synthesized in response to cell growth signals and helps to coordinate the progression of the cell cycle through the different stages, including DNA replication and cell division. It is expressed primarily in cells that are actively dividing, such as those in the liver, kidney, and testes. In the medical field, cyclin A1 is often studied in the context of cancer, as its overexpression has been linked to the development and progression of certain types of tumors.
Cyclins are a family of proteins that play a critical role in regulating the progression of the cell cycle in eukaryotic cells. They are synthesized and degraded in a cyclic manner, hence their name, and their levels fluctuate throughout the cell cycle. Cyclins interact with cyclin-dependent kinases (CDKs) to form cyclin-CDK complexes, which are responsible for phosphorylating target proteins and regulating cell cycle progression. Different cyclins are associated with different stages of the cell cycle, and their activity is tightly regulated by various mechanisms, including post-translational modifications and proteolysis. Dysregulation of cyclin expression or activity has been implicated in a variety of diseases, including cancer, where it is often associated with uncontrolled cell proliferation and tumor growth. Therefore, understanding the mechanisms that regulate cyclin expression and activity is important for developing new therapeutic strategies for cancer and other diseases.
Cyclin A2 is a protein that plays a role in regulating the cell cycle, which is the process by which cells grow, divide, and replicate their genetic material. Cyclin A2 is synthesized in response to cell growth signals and helps to coordinate the progression of the cell cycle through its interaction with cyclin-dependent kinases (CDKs). Specifically, Cyclin A2 forms a complex with CDK2, which is a key regulator of the G1/S transition, the point at which cells move from the G1 phase (resting phase) to the S phase (synthesis phase) of the cell cycle. This complex helps to phosphorylate and activate other proteins involved in the cell cycle, allowing cells to progress through the G1/S transition and enter the S phase. Cyclin A2 is also involved in the regulation of DNA replication and mitosis, the process by which cells divide into two daughter cells.
Cyclin-dependent kinase 4 (CDK4) is a protein that plays a critical role in regulating the cell cycle, which is the process by which cells divide and replicate. CDK4 is a member of the cyclin-dependent kinase (CDK) family of proteins, which are involved in regulating various cellular processes, including cell division, DNA replication, and transcription. CDK4 is activated by binding to cyclin D, a regulatory protein that is produced in response to growth signals. Once activated, CDK4 phosphorylates a number of target proteins, including the retinoblastoma protein (Rb), which is a key regulator of the cell cycle. Phosphorylation of Rb leads to its inactivation, allowing the cell to progress through the cell cycle and divide. Abnormal regulation of CDK4 activity has been implicated in a number of diseases, including cancer. For example, mutations in the CDK4 gene or overexpression of CDK4 have been found in various types of cancer, including breast, prostate, and lung cancer. In these cases, CDK4 may contribute to uncontrolled cell division and the development of tumors. In the medical field, CDK4 inhibitors are being developed as potential treatments for cancer. These drugs work by blocking the activity of CDK4, thereby inhibiting the growth and proliferation of cancer cells. Some CDK4 inhibitors have already been approved for use in certain types of cancer, and others are currently being tested in clinical trials.
Cyclin-dependent kinases (CDKs) are a family of protein kinases that play a critical role in regulating cell cycle progression in eukaryotic cells. They are activated by binding to specific regulatory proteins called cyclins, which are synthesized and degraded in a cyclic manner throughout the cell cycle. CDKs phosphorylate target proteins, including other kinases and transcription factors, to promote or inhibit cell cycle progression at specific points. Dysregulation of CDK activity has been implicated in a variety of diseases, including cancer, and is a target for therapeutic intervention.
Cyclin G1 is a protein that plays a role in regulating the cell cycle, which is the process by which cells grow, divide, and replicate their genetic material. Cyclin G1 is expressed at low levels in most cells, but its levels increase during the G1 phase of the cell cycle, which is the phase when the cell prepares for DNA replication. Cyclin G1 works by binding to and activating an enzyme called cyclin-dependent kinase 4 (CDK4), which in turn phosphorylates and inactivates the retinoblastoma protein (Rb). This inactivation of Rb allows the cell to progress through the G1 phase and enter the S phase, where DNA replication occurs. Dysregulation of cyclin G1 expression or activity has been implicated in the development of various types of cancer.
Cyclin G is a protein that plays a role in regulating the cell cycle, which is the process by which cells divide and grow. It is a type of cyclin, which are proteins that are involved in regulating the progression of the cell cycle through different phases. Cyclin G is expressed at low levels in most cells, but its levels increase during certain stages of the cell cycle, particularly during the G1 phase, which is the first phase of the cell cycle. Cyclin G is thought to help regulate the progression of the cell cycle by interacting with and activating cyclin-dependent kinases (CDKs), which are enzymes that control the progression of the cell cycle. Dysregulation of cyclin G expression or function has been implicated in the development of various types of cancer.
Cyclin-dependent kinase 2 (CDK2) is an enzyme that plays a critical role in cell cycle regulation. It is a member of the cyclin-dependent kinase (CDK) family of proteins, which are involved in the control of cell division and progression through the cell cycle. CDK2 is activated by binding to cyclin A, a regulatory protein that is expressed during the S phase of the cell cycle. Once activated, CDK2 phosphorylates a variety of target proteins, including the retinoblastoma protein (Rb), which is a key regulator of the cell cycle. Phosphorylation of Rb leads to its inactivation and the release of the transcription factor E2F, which promotes the transcription of genes required for DNA replication and cell division. CDK2 is also involved in the regulation of other cellular processes, including DNA repair, apoptosis, and differentiation. Dysregulation of CDK2 activity has been implicated in a number of diseases, including cancer, where it is often overexpressed or mutated. As such, CDK2 is a target for the development of new cancer therapies.
Cyclin C is a protein that plays a role in regulating the cell cycle, which is the process by which cells divide and grow. It is a member of the cyclin family of proteins, which are involved in regulating the progression of the cell cycle through different phases. Cyclin C is primarily expressed in the brain and is involved in the regulation of neural development and function. It has also been implicated in the development of certain types of cancer, including breast cancer and glioblastoma. In the medical field, cyclin C is studied as a potential target for the development of new treatments for these and other diseases.
Retinoblastoma protein (pRb) is a tumor suppressor protein that plays a critical role in regulating cell cycle progression and preventing the development of cancer. It is encoded by the RB1 gene, which is located on chromosome 13. In normal cells, pRb functions as a regulator of the cell cycle by binding to and inhibiting the activity of the E2F family of transcription factors. When cells are damaged or under stress, pRb is phosphorylated, which leads to its release from E2F and allows the cell to proceed through the cell cycle and divide. However, in cells with a mutated RB1 gene, pRb is unable to function properly, leading to uncontrolled cell division and the formation of tumors. Retinoblastoma is a type of eye cancer that occurs almost exclusively in children and is caused by mutations in the RB1 gene. Other types of cancer, such as osteosarcoma and small cell lung cancer, can also be associated with mutations in the RB1 gene.
CDC2-CDC28 kinases are a family of protein kinases that play a critical role in regulating cell cycle progression in eukaryotic cells. These kinases are named after the two genes that were originally identified in yeast, CDC2 and CDC28. CDC2-CDC28 kinases are involved in several key events during the cell cycle, including the initiation of DNA replication, the progression through the G1, S, G2, and M phases, and the regulation of mitosis. They are also involved in the regulation of cell growth, differentiation, and apoptosis. Inactivation of CDC2-CDC28 kinases can lead to cell cycle arrest, which can have both positive and negative effects on cell function. For example, cell cycle arrest can prevent the proliferation of cancer cells, but it can also lead to cell death in cells that are unable to repair damaged DNA. In the medical field, CDC2-CDC28 kinases are of interest as potential therapeutic targets for the treatment of various diseases, including cancer, as well as for the development of new drugs to regulate cell cycle progression and cell growth.
Cyclin B2 is a protein that plays a crucial role in regulating the progression of the cell cycle, particularly during the G2/M phase. It is a member of the cyclin family of proteins, which are involved in regulating the cell cycle by interacting with cyclin-dependent kinases (CDKs). Cyclin B2 is synthesized and degraded in a tightly regulated manner during the cell cycle. It is synthesized during the G2 phase and accumulates in the cell until the onset of mitosis, at which point it binds to and activates CDK1, forming the cyclin B1/CDK1 complex. This complex is essential for the initiation of mitosis and the proper progression of the cell through the M phase. Disruptions in the regulation of cyclin B2 expression or activity have been implicated in a variety of diseases, including cancer. For example, overexpression of cyclin B2 has been observed in several types of cancer, and it has been suggested that this may contribute to the uncontrolled proliferation of cancer cells. Conversely, loss of cyclin B2 function has been associated with defects in cell cycle progression and may contribute to the development of certain types of cancer.
Cyclin T is a protein that plays a role in regulating the progression of the cell cycle. It is a subunit of the cyclin-dependent kinase 9 (CDK9) complex, which is involved in the transcription of RNA. Cyclin T is essential for the activation of the transcription factor elongation factor 2 (EF2), which is responsible for the synthesis of proteins. In the context of the medical field, cyclin T has been implicated in the regulation of various cellular processes, including cell proliferation, differentiation, and apoptosis. Dysregulation of cyclin T has been associated with several diseases, including cancer, viral infections, and neurological disorders.
Oncogenes are genes that have the potential to cause cancer when they are mutated or expressed at high levels. Oncogenes are also known as proto-oncogenes, and they are involved in regulating cell growth and division. When oncogenes are mutated or expressed at high levels, they can cause uncontrolled cell growth and division, leading to the development of cancer. Oncogene proteins are the proteins that are produced by oncogenes. These proteins can play a variety of roles in the development and progression of cancer, including promoting cell growth and division, inhibiting cell death, and contributing to the formation of tumors.
Cyclin H is a protein that plays a role in the regulation of cell division. It is a component of the cyclin-dependent kinase (CDK) complex, which is responsible for phosphorylating target proteins and regulating the progression of the cell cycle. Cyclin H is involved in the transition from the G1 phase to the S phase of the cell cycle, where DNA replication occurs. It is also involved in the regulation of DNA repair and the maintenance of genomic stability. Mutations in the gene encoding cyclin H have been associated with an increased risk of certain types of cancer, including colorectal and ovarian cancer.
Cell cycle proteins are a group of proteins that play a crucial role in regulating the progression of the cell cycle. The cell cycle is a series of events that a cell goes through in order to divide and produce two daughter cells. It consists of four main phases: G1 (Gap 1), S (Synthesis), G2 (Gap 2), and M (Mitosis). Cell cycle proteins are involved in regulating the progression of each phase of the cell cycle, ensuring that the cell divides correctly and that the daughter cells have the correct number of chromosomes. Some of the key cell cycle proteins include cyclins, cyclin-dependent kinases (CDKs), and checkpoint proteins. Cyclins are proteins that are synthesized and degraded in a cyclic manner throughout the cell cycle. They bind to CDKs, which are enzymes that regulate cell cycle progression by phosphorylating target proteins. The activity of CDKs is tightly regulated by cyclins, ensuring that the cell cycle progresses in a controlled manner. Checkpoint proteins are proteins that monitor the cell cycle and ensure that the cell does not proceed to the next phase until all the necessary conditions are met. If any errors are detected, checkpoint proteins can halt the cell cycle and activate repair mechanisms to correct the problem. Overall, cell cycle proteins play a critical role in maintaining the integrity of the cell cycle and ensuring that cells divide correctly. Disruptions in the regulation of cell cycle proteins can lead to a variety of diseases, including cancer.
Cyclin G2 is a protein that plays a role in regulating the cell cycle, which is the process by which cells grow, divide, and replicate their genetic material. Cyclin G2 is involved in the transition from the G1 phase (the first stage of the cell cycle) to the S phase (the stage where DNA replication occurs). It is also involved in the regulation of the G2/M transition, which is the stage where the cell prepares to divide. In the medical field, Cyclin G2 has been implicated in the development of certain types of cancer, including breast cancer and ovarian cancer. It is also being studied as a potential target for cancer therapy.
CDC2 Protein Kinase is a type of enzyme that plays a crucial role in cell division and the regulation of the cell cycle. It is a serine/threonine protein kinase that is activated during the G2 phase of the cell cycle and is responsible for the initiation of mitosis. CDC2 is also involved in the regulation of DNA replication and the maintenance of genomic stability. In the medical field, CDC2 Protein Kinase is often studied in the context of cancer research, as its dysregulation has been linked to the development and progression of various types of cancer.
Cyclin-dependent kinase inhibitor p21 (p21) is a protein that plays a role in regulating the cell cycle, which is the process by which cells divide and grow. It is encoded by the CDKN1A gene and is a member of the Cip/Kip family of cyclin-dependent kinase inhibitors. In the cell cycle, the progression from one phase to the next is controlled by a series of checkpoints that ensure that the cell is ready to proceed. One of the key regulators of these checkpoints is the cyclin-dependent kinase (CDK) family of enzymes. CDKs are activated by binding to cyclins, which are proteins that are synthesized and degraded in a cyclic manner throughout the cell cycle. p21 acts as a CDK inhibitor by binding to and inhibiting the activity of cyclin-CDK complexes. This prevents the complexes from phosphorylating target proteins that are required for the progression of the cell cycle. As a result, p21 helps to prevent the cell from dividing when it is not ready, and it plays a role in preventing the development of cancer. In addition to its role in regulating the cell cycle, p21 has been implicated in a number of other cellular processes, including DNA repair, senescence, and apoptosis (programmed cell death). It is also involved in the response of cells to various stressors, such as DNA damage, oxidative stress, and hypoxia.
Protein-Serine-Threonine Kinases (PSTKs) are a family of enzymes that play a crucial role in regulating various cellular processes, including cell growth, differentiation, metabolism, and apoptosis. These enzymes phosphorylate specific amino acids, such as serine and threonine, on target proteins, thereby altering their activity, stability, or localization within the cell. PSTKs are involved in a wide range of diseases, including cancer, diabetes, cardiovascular disease, and neurodegenerative disorders. Therefore, understanding the function and regulation of PSTKs is important for developing new therapeutic strategies for these diseases.
Cyclin-dependent kinase inhibitor p27 (p27Kip1) is a protein that plays a role in regulating cell cycle progression. It is a member of the Cip/Kip family of cyclin-dependent kinase inhibitors, which also includes p21 and p57. In the cell cycle, the progression from one phase to the next is tightly regulated by a series of events that involve the activity of cyclin-dependent kinases (CDKs). CDKs are enzymes that are activated by binding to specific cyclins, which are proteins that are synthesized and degraded in a cyclic manner throughout the cell cycle. When CDKs are activated, they phosphorylate target proteins, which can either promote or inhibit cell cycle progression. p27Kip1 acts as a CDK inhibitor by binding to and inhibiting the activity of CDKs. It is primarily expressed in cells that are in a non-dividing state, such as terminally differentiated cells and quiescent cells. In these cells, p27Kip1 helps to maintain the cell in a non-dividing state by inhibiting the activity of CDKs, which prevents the cell from entering the cell cycle. In contrast, p27Kip1 is downregulated or lost in many types of cancer cells, where it is often associated with increased cell proliferation and tumor growth. This suggests that p27Kip1 may play a role in the development and progression of cancer.
Proto-oncogenes are normal genes that are involved in regulating cell growth and division. When these genes are mutated or overexpressed, they can become oncogenes, which can lead to the development of cancer. Proto-oncogenes are also known as proto-oncogene proteins.
Tumor suppressor proteins are a group of proteins that play a crucial role in regulating cell growth and preventing the development of cancer. These proteins act as brakes on the cell cycle, preventing cells from dividing and multiplying uncontrollably. They also help to repair damaged DNA and prevent the formation of tumors. Tumor suppressor proteins are encoded by genes that are located on specific chromosomes. When these genes are functioning properly, they produce proteins that help to regulate cell growth and prevent the development of cancer. However, when these genes are mutated or damaged, the proteins they produce may not function properly, leading to uncontrolled cell growth and the development of cancer. There are many different tumor suppressor proteins, each with its own specific function. Some of the most well-known tumor suppressor proteins include p53, BRCA1, and BRCA2. These proteins are involved in regulating cell cycle checkpoints, repairing damaged DNA, and preventing the formation of tumors. In summary, tumor suppressor proteins are a group of proteins that play a critical role in regulating cell growth and preventing the development of cancer. When these proteins are functioning properly, they help to maintain the normal balance of cell growth and division, but when they are mutated or damaged, they can contribute to the development of cancer.
Cyclin I is a protein that plays a role in regulating the cell cycle, which is the process by which cells grow, divide, and replicate their genetic material. Cyclin I is a type of cyclin, which are proteins that bind to and activate cyclin-dependent kinases (CDKs), enzymes that control the progression of the cell cycle. Cyclin I is involved in the G1 phase of the cell cycle, which is the first phase of the cycle and is characterized by cell growth and preparation for DNA replication. During the G1 phase, cyclin I helps to activate CDK4 and CDK6, which in turn phosphorylate and activate other proteins that are necessary for the progression of the cell cycle. Disruptions in the regulation of cyclin I and CDKs can lead to uncontrolled cell growth and the development of cancer.
In the medical field, RNA, Messenger (mRNA) refers to a type of RNA molecule that carries genetic information from DNA in the nucleus of a cell to the ribosomes, where proteins are synthesized. During the process of transcription, the DNA sequence of a gene is copied into a complementary RNA sequence called messenger RNA (mRNA). This mRNA molecule then leaves the nucleus and travels to the cytoplasm of the cell, where it binds to ribosomes and serves as a template for the synthesis of a specific protein. The sequence of nucleotides in the mRNA molecule determines the sequence of amino acids in the protein that is synthesized. Therefore, changes in the sequence of nucleotides in the mRNA molecule can result in changes in the amino acid sequence of the protein, which can affect the function of the protein and potentially lead to disease. mRNA molecules are often used in medical research and therapy as a way to introduce new genetic information into cells. For example, mRNA vaccines work by introducing a small piece of mRNA that encodes for a specific protein, which triggers an immune response in the body.
Lymphoma, Mantle-Cell is a type of non-Hodgkin's lymphoma, which is a cancer that affects the lymphatic system. The lymphatic system is a part of the immune system that helps to fight infections and diseases. Mantle-cell lymphoma typically affects the lymph nodes in the neck, armpits, and groin, but it can also affect other parts of the body, such as the spleen, liver, and bone marrow. Mantle-cell lymphoma is a relatively rare type of lymphoma, accounting for about 5% of all cases of non-Hodgkin's lymphoma. It is more common in older adults, with an average age of diagnosis of around 65 years. The exact cause of mantle-cell lymphoma is not known, but it is believed to be related to genetic changes in the cells of the immune system. Treatment options for mantle-cell lymphoma include chemotherapy, radiation therapy, and targeted therapy. In some cases, a stem cell transplant may also be recommended. The prognosis for mantle-cell lymphoma varies depending on the stage of the disease and the response to treatment.
Cyclin-dependent kinase inhibitor p16, also known as CDKN2A or p16INK4a, is a protein that plays a crucial role in regulating the cell cycle and preventing uncontrolled cell growth. It is encoded by the CDKN2A gene and is a member of the cyclin-dependent kinase inhibitor (CKI) family. In normal cells, p16 is expressed in response to DNA damage and acts as a brake on the cell cycle by inhibiting the activity of cyclin-dependent kinases (CDKs), which are enzymes that control cell cycle progression. When cells are damaged, p16 is activated and binds to CDK4 and CDK6, preventing them from phosphorylating and activating the retinoblastoma protein (Rb), which is a key regulator of the cell cycle. However, in many types of cancer, the CDKN2A gene is mutated or deleted, leading to a loss of p16 expression and allowing cells to bypass the cell cycle checkpoint controlled by p16. This can result in uncontrolled cell growth and the development of tumors. Therefore, p16 is considered a tumor suppressor gene, and its loss of function is associated with an increased risk of developing various types of cancer, including melanoma, lung cancer, and pancreatic cancer. In addition, p16 is also used as a diagnostic and prognostic marker in cancer, as its expression levels can be used to predict the aggressiveness of tumors and the response to treatment.
Proliferating Cell Nuclear Antigen (PCNA) is a protein that plays a crucial role in DNA replication and repair in cells. It is also known as Replication Factor C (RFC) subunit 4 or proliferating cell nuclear antigen-like 1 (PCNA-like 1). PCNA is a highly conserved protein that is found in all eukaryotic cells. It is a homotrimeric protein, meaning that it is composed of three identical subunits. Each subunit has a central channel that can bind to DNA, and it is this channel that is responsible for the interaction of PCNA with other proteins involved in DNA replication and repair. During DNA replication, PCNA forms a complex with other proteins, including DNA polymerase δ and the replication factor C (RFC) complex. This complex is responsible for unwinding the DNA double helix, synthesizing new DNA strands, and ensuring that the newly synthesized strands are correctly paired with the template strands. PCNA is also involved in DNA repair processes, particularly in the repair of DNA damage caused by ultraviolet (UV) radiation. In this context, PCNA interacts with other proteins, such as the X-ray repair cross-complementing protein 1 (XRCC1), to facilitate the repair of DNA damage. Overall, PCNA is a critical protein in the maintenance of genomic stability and the prevention of DNA damage-induced diseases, such as cancer.
E2F transcription factors are a family of proteins that play a critical role in regulating the cell cycle and controlling cell proliferation. They are named for their ability to bind to the E2 promoter region of genes that are involved in cell cycle progression. There are six known E2F transcription factors in humans, which are classified into three groups: E2F1-3, DP1-3, and E4F1. E2F1-3 are primarily involved in regulating cell cycle progression, while DP1-3 are required for the formation of stable E2F-DP complexes that are necessary for transcriptional activation. E4F1 is a transcriptional repressor that is involved in regulating DNA repair and cell death. E2F transcription factors are activated by the binding of cyclin-dependent kinases (CDKs) to cyclins, which occur during the G1 phase of the cell cycle. Once activated, E2F transcription factors bind to specific DNA sequences and promote the transcription of genes involved in cell cycle progression, such as those encoding cyclins and other cell cycle regulators. Abnormal regulation of E2F transcription factors has been implicated in a variety of human diseases, including cancer. For example, overexpression of E2F1 has been associated with the development of several types of cancer, including breast, lung, and ovarian cancer. Conversely, loss of E2F1 function has been shown to inhibit tumor growth and improve the efficacy of cancer therapies.
Beta-catenin is a protein that plays a crucial role in the regulation of cell adhesion and signaling pathways in the body. In the medical field, beta-catenin is often studied in the context of cancer, as mutations in the beta-catenin gene (CTNNB1) can lead to the development of various types of cancer, including colorectal cancer, endometrial cancer, and ovarian cancer. In normal cells, beta-catenin is a component of the cadherin adhesion complex, which helps cells stick together and maintain tissue integrity. However, in cancer cells, mutations in the beta-catenin gene can lead to the accumulation of beta-catenin in the cytoplasm and nucleus, where it can activate downstream signaling pathways that promote cell proliferation and survival. Beta-catenin is also involved in the regulation of other cellular processes, such as cell migration, differentiation, and apoptosis. As such, it is a potential target for the development of new cancer therapies.
Cyclin-dependent kinase 6 (CDK6) is an enzyme that plays a critical role in cell cycle regulation. It is a member of the cyclin-dependent kinase (CDK) family, which are regulatory enzymes that control the progression of cells through the different phases of the cell cycle. CDK6 is activated by binding to cyclin D, a regulatory protein that is expressed during the G1 phase of the cell cycle. Once activated, CDK6 phosphorylates a number of target proteins, including the retinoblastoma protein (Rb), which is a key regulator of the G1/S transition. Phosphorylation of Rb leads to its inactivation, allowing the cell to progress through the G1 phase and enter the S phase of the cell cycle. CDK6 is also involved in the regulation of other cellular processes, including DNA replication, transcription, and cell proliferation. Dysregulation of CDK6 activity has been implicated in the development of a number of human diseases, including cancer. For example, overexpression of CDK6 has been observed in many types of cancer, and it is thought to contribute to the uncontrolled cell proliferation that characterizes these diseases.
Transcription factors are proteins that regulate gene expression by binding to specific DNA sequences and controlling the transcription of genetic information from DNA to RNA. They play a crucial role in the development and function of cells and tissues in the body. In the medical field, transcription factors are often studied as potential targets for the treatment of diseases such as cancer, where their activity is often dysregulated. For example, some transcription factors are overexpressed in certain types of cancer cells, and inhibiting their activity may help to slow or stop the growth of these cells. Transcription factors are also important in the development of stem cells, which have the ability to differentiate into a wide variety of cell types. By understanding how transcription factors regulate gene expression in stem cells, researchers may be able to develop new therapies for diseases such as diabetes and heart disease. Overall, transcription factors are a critical component of gene regulation and have important implications for the development and treatment of many diseases.
Glycogen Synthase Kinase 3 (GSK3) is a family of serine/threonine protein kinases that play a crucial role in various cellular processes, including metabolism, cell signaling, and gene expression. In the medical field, GSK3 has been implicated in the development and progression of several diseases, including diabetes, neurodegenerative disorders, and cancer. GSK3 is activated by various stimuli, including stress, inflammation, and insulin resistance, and its activity is regulated by phosphorylation and dephosphorylation. When activated, GSK3 phosphorylates and inactivates glycogen synthase, the enzyme responsible for glycogen synthesis, leading to reduced glycogen storage in the liver and muscles. This can contribute to the development of diabetes and other metabolic disorders. In addition to its role in metabolism, GSK3 has also been implicated in the regulation of cell signaling pathways, including the Wnt signaling pathway, which plays a critical role in cell proliferation, differentiation, and survival. Dysregulation of GSK3 activity in the Wnt signaling pathway has been implicated in the development of several types of cancer, including colon, breast, and ovarian cancer. Overall, GSK3 is a key regulator of cellular processes and its dysregulation has been implicated in the development and progression of several diseases. As such, it is an important target for the development of new therapeutic strategies for these diseases.
Ki-67 is a protein found in the nuclei of cells that are actively dividing. It is a useful marker for assessing the growth rate of tumors and is often used in conjunction with other markers to help diagnose and predict the behavior of cancer. The Ki-67 antigen is named after the Danish pathologist, Kai Erik Nielsen, who first described it in the 1980s. It is typically measured using immunohistochemistry, a technique that uses antibodies to detect specific proteins in tissue samples.
Cell transformation, neoplastic refers to the process by which normal cells in the body undergo genetic changes that cause them to become cancerous or malignant. This process involves the accumulation of mutations in genes that regulate cell growth, division, and death, leading to uncontrolled cell proliferation and the formation of tumors. Neoplastic transformation can occur in any type of cell in the body, and it can be caused by a variety of factors, including exposure to carcinogens, radiation, viruses, and inherited genetic mutations. Once a cell has undergone neoplastic transformation, it can continue to divide and grow uncontrollably, invading nearby tissues and spreading to other parts of the body through the bloodstream or lymphatic system. The diagnosis of neoplastic transformation typically involves a combination of clinical examination, imaging studies, and biopsy. Treatment options for neoplastic transformation depend on the type and stage of cancer, as well as the patient's overall health and preferences. Common treatments include surgery, radiation therapy, chemotherapy, targeted therapy, and immunotherapy.
Breast neoplasms refer to abnormal growths or tumors in the breast tissue. These growths can be benign (non-cancerous) or malignant (cancerous). Benign breast neoplasms are usually not life-threatening, but they can cause discomfort or cosmetic concerns. Malignant breast neoplasms, on the other hand, can spread to other parts of the body and are considered a serious health threat. Some common types of breast neoplasms include fibroadenomas, ductal carcinoma in situ (DCIS), invasive ductal carcinoma, and invasive lobular carcinoma.
F-box proteins are a family of proteins that play a role in the regulation of protein degradation in cells. They are involved in the ubiquitin-proteasome pathway, which is the primary mechanism by which cells degrade and recycle proteins. F-box proteins are characterized by an F-box domain, which is a protein-protein interaction module that binds to other proteins, often through their ubiquitin modification. F-box proteins are often components of larger protein complexes, such as the SCF (Skp1-Cullin-F-box) complex, which is involved in the degradation of specific target proteins. Dysregulation of F-box proteins has been implicated in a number of diseases, including cancer, neurodegenerative disorders, and developmental disorders.
Tumor suppressor protein p53 is a protein that plays a crucial role in regulating cell growth and preventing the development of cancer. It is encoded by the TP53 gene and is one of the most commonly mutated genes in human cancer. The p53 protein acts as a "guardian of the genome" by detecting DNA damage and initiating a series of cellular responses to repair the damage or trigger programmed cell death (apoptosis) if the damage is too severe. This helps to prevent the accumulation of mutations in the DNA that can lead to the development of cancer. In addition to its role in preventing cancer, p53 also plays a role in regulating cell cycle progression, DNA repair, and the response to cellular stress. Mutations in the TP53 gene can lead to the production of a non-functional or mutated p53 protein, which can result in the loss of these important functions and contribute to the development of cancer. Overall, the p53 protein is a critical regulator of cell growth and survival, and its dysfunction is a common feature of many types of cancer.
DNA-binding proteins are a class of proteins that interact with DNA molecules to regulate gene expression. These proteins recognize specific DNA sequences and bind to them, thereby affecting the transcription of genes into messenger RNA (mRNA) and ultimately the production of proteins. DNA-binding proteins play a crucial role in many biological processes, including cell division, differentiation, and development. They can act as activators or repressors of gene expression, depending on the specific DNA sequence they bind to and the cellular context in which they are expressed. Examples of DNA-binding proteins include transcription factors, histones, and non-histone chromosomal proteins. Transcription factors are proteins that bind to specific DNA sequences and regulate the transcription of genes by recruiting RNA polymerase and other factors to the promoter region of a gene. Histones are proteins that package DNA into chromatin, and non-histone chromosomal proteins help to organize and regulate chromatin structure. DNA-binding proteins are important targets for drug discovery and development, as they play a central role in many diseases, including cancer, genetic disorders, and infectious diseases.
RNA, Small Interfering (siRNA) is a type of non-coding RNA molecule that plays a role in gene regulation. siRNA is approximately 21-25 nucleotides in length and is derived from double-stranded RNA (dsRNA) molecules. In the medical field, siRNA is used as a tool for gene silencing, which involves inhibiting the expression of specific genes. This is achieved by introducing siRNA molecules that are complementary to the target mRNA sequence, leading to the degradation of the mRNA and subsequent inhibition of protein synthesis. siRNA has potential applications in the treatment of various diseases, including cancer, viral infections, and genetic disorders. It is also used in research to study gene function and regulation. However, the use of siRNA in medicine is still in its early stages, and there are several challenges that need to be addressed before it can be widely used in clinical practice.
Proto-oncogene proteins c-myc is a family of proteins that play a role in regulating cell growth and division. They are also known as myc proteins. The c-myc protein is encoded by the MYC gene, which is located on chromosome 8. The c-myc protein is a transcription factor, which means that it helps to regulate the expression of other genes. When the c-myc protein is overexpressed or mutated, it can contribute to the development of cancer. In normal cells, the c-myc protein helps to control the cell cycle and prevent uncontrolled cell growth. However, in cancer cells, the c-myc protein may be overactive or mutated, leading to uncontrolled cell growth and the formation of tumors.
E2F1 transcription factor is a protein that plays a crucial role in regulating the cell cycle and cell proliferation. It is a member of the E2F family of transcription factors, which are involved in controlling the expression of genes that are necessary for cell cycle progression and DNA replication. E2F1 is activated during the G1 phase of the cell cycle, when the cell is preparing to divide. It binds to specific DNA sequences in the promoter regions of target genes, such as those involved in DNA replication and cell cycle progression, and promotes their transcription. In this way, E2F1 helps to coordinate the various events that occur during the cell cycle and ensure that the cell divides properly. Abnormal regulation of E2F1 has been implicated in a number of diseases, including cancer. For example, overexpression of E2F1 has been observed in many types of cancer, and it is thought to contribute to the uncontrolled proliferation of cancer cells. Conversely, loss of E2F1 function has been associated with impaired cell cycle progression and reduced cell proliferation, which may contribute to the development of certain types of cancer. Overall, E2F1 transcription factor plays a critical role in regulating the cell cycle and cell proliferation, and its dysregulation has been implicated in a number of diseases, including cancer.
Nuclear proteins are proteins that are found within the nucleus of a cell. The nucleus is the control center of the cell, where genetic material is stored and regulated. Nuclear proteins play a crucial role in many cellular processes, including DNA replication, transcription, and gene regulation. There are many different types of nuclear proteins, each with its own specific function. Some nuclear proteins are involved in the structure and organization of the nucleus itself, while others are involved in the regulation of gene expression. Nuclear proteins can also interact with other proteins, DNA, and RNA molecules to carry out their functions. In the medical field, nuclear proteins are often studied in the context of diseases such as cancer, where changes in the expression or function of nuclear proteins can contribute to the development and progression of the disease. Additionally, nuclear proteins are important targets for drug development, as they can be targeted to treat a variety of diseases.
Transcription factor DP1 is a protein that plays a role in regulating gene expression. It is a member of the basic helix-loop-helix (bHLH) family of transcription factors, which are proteins that bind to specific DNA sequences and help to control the transcription of genes. DP1 is encoded by the "DPF1" gene, which is located on chromosome 12 in humans. DP1 is involved in the development and differentiation of various cell types, including neurons, muscle cells, and immune cells. It has been implicated in a number of different diseases, including cancer, neurological disorders, and autoimmune diseases. For example, mutations in the "DPF1" gene have been associated with an increased risk of developing certain types of cancer, such as breast cancer and ovarian cancer. Additionally, DP1 has been shown to play a role in the development of multiple sclerosis, an autoimmune disorder that affects the central nervous system.
Retinoblastoma-like protein p107 (p107) is a protein that plays a role in regulating cell cycle progression and cell differentiation. It is a member of the retinoblastoma protein family, which includes other proteins such as pRb and p130. Like other members of this family, p107 is involved in the regulation of the G1/S transition of the cell cycle, which is the process by which cells prepare to divide. It does this by binding to specific DNA sequences and inhibiting the activity of proteins that promote cell cycle progression. In addition to its role in cell cycle regulation, p107 has also been implicated in the regulation of cell differentiation and the development of certain types of cancer.
CDC25 phosphatases are a family of enzymes that play a critical role in regulating cell cycle progression in eukaryotic cells. These enzymes are named after the cell division cycle 25 (CDC25) gene family, which encodes for the phosphatases. CDC25 phosphatases are responsible for dephosphorylating tyrosine residues on cyclin-dependent kinases (CDKs), which are key regulators of cell cycle progression. By removing phosphate groups from CDKs, CDC25 phosphatases activate these enzymes, allowing them to phosphorylate and activate other proteins involved in cell cycle progression. In addition to their role in cell cycle regulation, CDC25 phosphatases have also been implicated in a variety of other cellular processes, including DNA repair, apoptosis, and cancer development. Dysregulation of CDC25 phosphatase activity has been linked to several types of cancer, including breast, ovarian, and colorectal cancer. Overall, CDC25 phosphatases are important regulators of cell cycle progression and have important implications for human health and disease.
Neoplasm proteins are proteins that are produced by cancer cells. These proteins are often abnormal and can contribute to the growth and spread of cancer. They can be detected in the blood or other body fluids, and their presence can be used as a diagnostic tool for cancer. Some neoplasm proteins are also being studied as potential targets for cancer treatment.
Retinoblastoma-Binding Protein 1 (RBBP1) is a protein that plays a role in the regulation of gene expression. It is encoded by the RBBP1 gene and is found in the nucleus of cells. RBBP1 is a component of the Retinoblastoma protein (pRb) pathway, which is involved in the regulation of the cell cycle and the prevention of uncontrolled cell growth. In the context of retinoblastoma, a type of eye cancer that occurs in children, RBBP1 is thought to play a role in the development and progression of the disease.
The proteasome endopeptidase complex is a large protein complex found in the cells of all eukaryotic organisms. It is responsible for breaking down and recycling damaged or unnecessary proteins within the cell. The proteasome is composed of two main subunits: the 20S core particle, which contains the proteolytic active sites, and the 19S regulatory particle, which recognizes and unfolds target proteins for degradation. The proteasome plays a critical role in maintaining cellular homeostasis and is involved in a wide range of cellular processes, including cell cycle regulation, immune response, and protein quality control. Dysregulation of the proteasome has been implicated in a number of diseases, including cancer, neurodegenerative disorders, and autoimmune diseases.
S-phase kinase-associated proteins (SKAPs) are a family of proteins that play a role in regulating the progression of the cell cycle, specifically during the S phase (DNA synthesis phase). They are involved in the regulation of DNA replication and repair, and are also implicated in the development of certain types of cancer. SKAPs are activated by the cyclin-dependent kinase (CDK) complex, which is a key regulator of the cell cycle. Dysregulation of SKAPs has been linked to various cellular processes, including cell proliferation, apoptosis, and differentiation.
In the medical field, "trans-activators" refer to proteins or molecules that activate the transcription of a gene, which is the process by which the information in a gene is used to produce a functional product, such as a protein. Trans-activators can bind to specific DNA sequences near a gene and recruit other proteins, such as RNA polymerase, to initiate transcription. They can also modify the chromatin structure around a gene to make it more accessible to transcription machinery. Trans-activators play important roles in regulating gene expression and are involved in many biological processes, including development, differentiation, and disease.
Recombinant fusion proteins are proteins that are produced by combining two or more genes in a single molecule. These proteins are typically created using genetic engineering techniques, such as recombinant DNA technology, to insert one or more genes into a host organism, such as bacteria or yeast, which then produces the fusion protein. Fusion proteins are often used in medical research and drug development because they can have unique properties that are not present in the individual proteins that make up the fusion. For example, a fusion protein might be designed to have increased stability, improved solubility, or enhanced targeting to specific cells or tissues. Recombinant fusion proteins have a wide range of applications in medicine, including as therapeutic agents, diagnostic tools, and research reagents. Some examples of recombinant fusion proteins used in medicine include antibodies, growth factors, and cytokines.
Proto-oncogene proteins c-akt, also known as protein kinase B (PKB), is a serine/threonine kinase that plays a critical role in various cellular processes, including cell survival, proliferation, and metabolism. It is a member of the Akt family of kinases, which are activated by various growth factors and cytokines. In the context of cancer, c-akt has been shown to be frequently activated in many types of tumors and is often associated with poor prognosis. Activation of c-akt can lead to increased cell survival and resistance to apoptosis, which can contribute to tumor growth and progression. Additionally, c-akt has been implicated in the regulation of angiogenesis, invasion, and metastasis, further contributing to the development and progression of cancer. Therefore, the study of c-akt and its role in cancer has become an important area of research in the medical field, with the goal of developing targeted therapies to inhibit its activity and potentially treat cancer.
DNA primers are short, single-stranded DNA molecules that are used in a variety of molecular biology techniques, including polymerase chain reaction (PCR) and DNA sequencing. They are designed to bind to specific regions of a DNA molecule, and are used to initiate the synthesis of new DNA strands. In PCR, DNA primers are used to amplify specific regions of DNA by providing a starting point for the polymerase enzyme to begin synthesizing new DNA strands. The primers are complementary to the target DNA sequence, and are added to the reaction mixture along with the DNA template, nucleotides, and polymerase enzyme. The polymerase enzyme uses the primers as a template to synthesize new DNA strands, which are then extended by the addition of more nucleotides. This process is repeated multiple times, resulting in the amplification of the target DNA sequence. DNA primers are also used in DNA sequencing to identify the order of nucleotides in a DNA molecule. In this application, the primers are designed to bind to specific regions of the DNA molecule, and are used to initiate the synthesis of short DNA fragments. The fragments are then sequenced using a variety of techniques, such as Sanger sequencing or next-generation sequencing. Overall, DNA primers are an important tool in molecular biology, and are used in a wide range of applications to study and manipulate DNA.
Protein kinases are enzymes that catalyze the transfer of a phosphate group from ATP (adenosine triphosphate) to specific amino acid residues on proteins. This process, known as phosphorylation, can alter the activity, localization, or stability of the target protein, and is a key mechanism for regulating many cellular processes, including cell growth, differentiation, metabolism, and signaling pathways. Protein kinases are classified into different families based on their sequence, structure, and substrate specificity. Some of the major families of protein kinases include serine/threonine kinases, tyrosine kinases, and dual-specificity kinases. Each family has its own unique functions and roles in cellular signaling. In the medical field, protein kinases are important targets for the development of drugs for the treatment of various diseases, including cancer, diabetes, and cardiovascular disease. Many cancer drugs target specific protein kinases that are overactive in cancer cells, while drugs for diabetes and cardiovascular disease often target kinases involved in glucose metabolism and blood vessel function, respectively.
Microtubule-associated proteins (MAPs) are a group of proteins that bind to microtubules, which are important components of the cytoskeleton in cells. These proteins play a crucial role in regulating the dynamics of microtubules, including their assembly, disassembly, and stability. MAPs are involved in a wide range of cellular processes, including cell division, intracellular transport, and the maintenance of cell shape. They can also play a role in the development of diseases such as cancer, where the abnormal regulation of microtubules and MAPs can contribute to the growth and spread of tumors. There are many different types of MAPs, each with its own specific functions and mechanisms of action. Some MAPs are involved in regulating the dynamics of microtubules, while others are involved in the transport of molecules along microtubules. Some MAPs are also involved in the organization and function of the mitotic spindle, which is essential for the proper segregation of chromosomes during cell division. Overall, MAPs are important regulators of microtubule dynamics and play a crucial role in many cellular processes. Understanding the function of these proteins is important for developing new treatments for diseases that are associated with abnormal microtubule regulation.
Protamine kinase is an enzyme that is involved in the regulation of blood clotting. It is responsible for converting protamine sulfate, a substance that is used to neutralize the anticoagulant effects of heparin, into protamine. Protamine sulfate is often used in conjunction with heparin during medical procedures, such as surgery or catheterization, to prevent excessive bleeding. Protamine kinase helps to ensure that the appropriate amount of protamine is present to neutralize the heparin, preventing the formation of blood clots.
In the medical field, carrier proteins are proteins that transport molecules across cell membranes or within cells. These proteins bind to specific molecules, such as hormones, nutrients, or waste products, and facilitate their movement across the membrane or within the cell. Carrier proteins play a crucial role in maintaining the proper balance of molecules within cells and between cells. They are involved in a wide range of physiological processes, including nutrient absorption, hormone regulation, and waste elimination. There are several types of carrier proteins, including facilitated diffusion carriers, active transport carriers, and ion channels. Each type of carrier protein has a specific function and mechanism of action. Understanding the role of carrier proteins in the body is important for diagnosing and treating various medical conditions, such as genetic disorders, metabolic disorders, and neurological disorders.
Cyclin-dependent kinase inhibitors (CDKIs) are a group of proteins that regulate the cell cycle by inhibiting the activity of cyclin-dependent kinases (CDKs). CDKs are a family of enzymes that play a critical role in regulating cell cycle progression by phosphorylating target proteins. CDKIs bind to CDKs and prevent them from phosphorylating their target proteins, thereby inhibiting cell cycle progression. CDKIs are important regulators of cell cycle progression and are involved in a variety of cellular processes, including DNA replication, chromosome segregation, and apoptosis. Dysregulation of CDKIs has been implicated in a number of diseases, including cancer, where the overexpression or loss of function of CDKIs can lead to uncontrolled cell proliferation and the development of tumors. CDKIs are classified into two main groups: the INK4 family (p16INK4a, p15INK4b, p18INK4c, and p19INK4d) and the Cip/Kip family (p21Cip1, p27Kip1, and p57Kip2). The INK4 family members are primarily involved in inhibiting CDK4 and CDK6, while the Cip/Kip family members can inhibit multiple CDKs.
Threonine is an essential amino acid that plays a crucial role in various biological processes in the human body. It is a polar amino acid with a hydroxyl group (-OH) attached to the alpha carbon atom, which makes it hydrophilic and capable of forming hydrogen bonds. In the medical field, threonine is important for several reasons. Firstly, it is a building block of proteins, which are essential for the structure and function of cells and tissues in the body. Secondly, threonine is involved in the metabolism of carbohydrates and lipids, which are important sources of energy for the body. Thirdly, threonine is a precursor for the synthesis of several important molecules, including carnitine, which plays a role in the metabolism of fatty acids. Threonine deficiency can lead to a range of health problems, including muscle wasting, impaired growth and development, and weakened immune function. It is therefore important to ensure that the body receives adequate amounts of threonine through a balanced diet or supplements.
Carcinoma, Squamous Cell is a type of cancer that originates in the squamous cells, which are thin, flat cells that line the surface of the body. Squamous cells are found in the skin, mouth, throat, lungs, and other organs. Carcinoma, Squamous Cell can develop in any part of the body where squamous cells are present, but it is most commonly found in the head and neck, lungs, and skin. The exact cause of Squamous Cell Carcinoma is not always clear, but it is often associated with exposure to certain substances, such as tobacco smoke, alcohol, and certain chemicals. It can also develop as a result of chronic inflammation or infection, such as HPV (human papillomavirus) infection in the cervix. Symptoms of Squamous Cell Carcinoma can vary depending on the location of the tumor, but may include a persistent sore or lesion that does not heal, a change in the appearance of the skin or mucous membranes, difficulty swallowing or breathing, and unexplained weight loss. Treatment for Squamous Cell Carcinoma typically involves surgery to remove the tumor, followed by radiation therapy or chemotherapy to kill any remaining cancer cells. In some cases, targeted therapy or immunotherapy may also be used. The prognosis for Squamous Cell Carcinoma depends on the stage of the cancer at the time of diagnosis and the overall health of the patient.
Phosphatidylinositol 3-kinases (PI3Ks) are a family of enzymes that play a critical role in cellular signaling pathways. They are involved in a wide range of cellular processes, including cell growth, proliferation, differentiation, survival, migration, and metabolism. PI3Ks are activated by various extracellular signals, such as growth factors, hormones, and neurotransmitters, and they generate second messengers by phosphorylating phosphatidylinositol lipids on the inner leaflet of the plasma membrane. This leads to the recruitment and activation of downstream effector molecules, such as protein kinases and phosphatases, which regulate various cellular processes. Dysregulation of PI3K signaling has been implicated in the development of various diseases, including cancer, diabetes, and neurological disorders. Therefore, PI3Ks are important targets for the development of therapeutic agents for these diseases.
Maturation-Promoting Factor (MPF) is a complex of proteins that plays a crucial role in the regulation of cell cycle progression and the initiation of mitosis in eukaryotic cells. It is composed of two subunits: cyclin B and cyclin-dependent kinase (CDK1). During the cell cycle, MPF is synthesized and activated during the G2 phase, and it remains active until the end of mitosis. MPF promotes the progression of the cell cycle by phosphorylating various target proteins, including the nuclear envelope, kinetochores, and other cell cycle regulators. MPF is also involved in the regulation of apoptosis, the process of programmed cell death. When cells are damaged or stressed, MPF can be activated to trigger apoptosis, which helps to eliminate damaged or abnormal cells. In the medical field, MPF is of interest because it plays a critical role in the development and progression of many diseases, including cancer. Abnormal regulation of MPF activity has been linked to the development of various types of cancer, and targeting MPF has been proposed as a potential therapeutic strategy for cancer treatment.
Mitogen-Activated Protein Kinases (MAPKs) are a family of enzymes that play a crucial role in cellular signaling pathways. They are involved in regulating various cellular processes such as cell growth, differentiation, proliferation, survival, and apoptosis. MAPKs are activated by extracellular signals such as growth factors, cytokines, and hormones, which bind to specific receptors on the cell surface. This activation leads to a cascade of phosphorylation events, where MAPKs phosphorylate and activate downstream effector molecules, such as transcription factors, that regulate gene expression. In the medical field, MAPKs are of great interest due to their involvement in various diseases, including cancer, inflammatory disorders, and neurological disorders. For example, mutations in MAPK signaling pathways are commonly found in many types of cancer, and targeting these pathways has become an important strategy for cancer therapy. Additionally, MAPKs are involved in the regulation of immune responses, and dysregulation of these pathways has been implicated in various inflammatory disorders. Finally, MAPKs play a role in the development and maintenance of the nervous system, and dysfunction of these pathways has been linked to neurological disorders such as Alzheimer's disease and Parkinson's disease.
Luciferases are enzymes that catalyze the oxidation of luciferin, a small molecule, to produce light. In the medical field, luciferases are commonly used as reporters in bioluminescence assays, which are used to measure gene expression, protein-protein interactions, and other biological processes. One of the most well-known examples of luciferases in medicine is the green fluorescent protein (GFP) luciferase, which is derived from the jellyfish Aequorea victoria. GFP luciferase is used in a variety of applications, including monitoring gene expression in living cells and tissues, tracking the movement of cells and proteins in vivo, and studying the dynamics of signaling pathways. Another example of a luciferase used in medicine is the firefly luciferase, which is derived from the firefly Photinus pyralis. Firefly luciferase is used in bioluminescence assays to measure the activity of various enzymes and to study the metabolism of drugs and other compounds. Overall, luciferases are valuable tools in the medical field because they allow researchers to visualize and quantify biological processes in a non-invasive and sensitive manner.
DNA, or deoxyribonucleic acid, is a molecule that carries genetic information in living organisms. It is composed of four types of nitrogen-containing molecules called nucleotides, which are arranged in a specific sequence to form the genetic code. In the medical field, DNA is often studied as a tool for understanding and diagnosing genetic disorders. Genetic disorders are caused by changes in the DNA sequence that can affect the function of genes, leading to a variety of health problems. By analyzing DNA, doctors and researchers can identify specific genetic mutations that may be responsible for a particular disorder, and develop targeted treatments or therapies to address the underlying cause of the condition. DNA is also used in forensic science to identify individuals based on their unique genetic fingerprint. This is because each person's DNA sequence is unique, and can be used to distinguish one individual from another. DNA analysis is also used in criminal investigations to help solve crimes by linking DNA evidence to suspects or victims.
Cyclin-dependent kinase 9 (CDK9) is an enzyme that plays a crucial role in regulating gene expression by phosphorylating the C-terminal domain (CTD) of RNA polymerase II (Pol II). CDK9 is a component of the positive transcription elongation factor b (P-TEFb) complex, which is responsible for promoting the transition of Pol II from initiation to elongation during transcription. CDK9 is activated by the cyclin T1 or cyclin T2 proteins, which bind to the kinase and stimulate its activity. The P-TEFb complex is involved in the regulation of many genes, including those involved in cell proliferation, differentiation, and survival. Dysregulation of CDK9 activity has been implicated in various diseases, including cancer, HIV infection, and neurological disorders. CDK9 inhibitors are being developed as potential therapeutic agents for the treatment of various diseases, including cancer and HIV infection. These inhibitors target the interaction between CDK9 and its cyclin partners, thereby inhibiting the activity of the P-TEFb complex and blocking transcription.
Bromodeoxyuridine (BrdU) is a synthetic analog of the nucleoside thymidine, which is a building block of DNA. It is commonly used in the medical field as a marker for DNA synthesis and cell proliferation. BrdU is incorporated into newly synthesized DNA during the S phase of the cell cycle, when DNA replication occurs. This makes it possible to detect cells that are actively dividing by staining for BrdU. BrdU staining is often used in immunohistochemistry and flow cytometry to study the proliferation of cells in various tissues and organs, including the brain, bone marrow, and skin. BrdU is also used in some cancer treatments, such as chemotherapy and radiation therapy, to target rapidly dividing cancer cells. By inhibiting DNA synthesis, BrdU can slow down or stop the growth of cancer cells, making them more susceptible to treatment. However, it is important to note that BrdU can also cause DNA damage and has been associated with an increased risk of cancer in some studies. Therefore, its use in medical research and treatment should be carefully monitored and regulated.
Ubiquitins are small, highly conserved proteins that are involved in a variety of cellular processes, including protein degradation, signal transduction, and gene expression. In the medical field, ubiquitins are often studied in the context of diseases such as cancer, neurodegenerative disorders, and autoimmune diseases. One of the key functions of ubiquitins is to mark proteins for degradation by the proteasome, a large protein complex that breaks down and removes damaged or unnecessary proteins from the cell. This process is essential for maintaining cellular homeostasis and regulating the levels of specific proteins in the cell. In addition to their role in protein degradation, ubiquitins are also involved in a number of other cellular processes, including cell cycle regulation, DNA repair, and immune response. Dysregulation of ubiquitin-mediated processes has been implicated in a variety of diseases, including cancer, where it can contribute to the development and progression of tumors. Overall, ubiquitins are an important class of proteins that play a critical role in many cellular processes, and their dysfunction can have significant consequences for human health.
E2F4 Transcription Factor is a protein that plays a role in regulating the cell cycle and cell proliferation. It is a member of the E2F family of transcription factors, which are involved in controlling the expression of genes that are necessary for cell division and growth. E2F4 is activated by the binding of other proteins, such as Cyclin E, and it in turn regulates the expression of genes that are necessary for the progression of the cell cycle. Dysregulation of E2F4 has been implicated in the development of various types of cancer, including breast, ovarian, and prostate cancer.
Ubiquitin-Protein Ligase Complexes (UPCs) are multi-protein complexes that play a crucial role in the process of protein degradation in cells. These complexes are responsible for attaching small protein molecules called ubiquitin to specific target proteins, which marks them for degradation by the proteasome, a large protein complex that breaks down proteins into smaller peptides. UPCs are composed of several subunits, including E1, E2, and E3 enzymes, which work together to transfer ubiquitin from one enzyme to another and ultimately to the target protein. The E1 enzyme activates ubiquitin, while the E2 enzyme binds to it and transfers it to the E3 enzyme, which recognizes the target protein and facilitates its ubiquitination. UPCs are involved in a wide range of cellular processes, including cell cycle regulation, DNA repair, and the regulation of protein levels. Dysregulation of UPCs has been implicated in several diseases, including cancer, neurodegenerative disorders, and autoimmune diseases. Therefore, understanding the function and regulation of UPCs is an important area of research in the medical field.
STAT3 (Signal Transducer and Activator of Transcription 3) is a transcription factor that plays a critical role in regulating gene expression in response to various signaling pathways, including cytokines, growth factors, and hormones. In the medical field, STAT3 is often studied in the context of cancer, as it is frequently activated in many types of tumors and is involved in promoting cell proliferation, survival, and invasion. Dysregulation of STAT3 signaling has been implicated in the development and progression of various cancers, including breast, prostate, and lung cancer. Additionally, STAT3 has been shown to play a role in other diseases, such as autoimmune disorders and inflammatory diseases. Targeting STAT3 signaling is therefore an active area of research in the development of new cancer therapies and other treatments.
Cyclin D1
Cyclin-dependent kinase
In situ lymphoid neoplasia
Mir-503 microRNA precursor family
MYBL2
STK11
MIR503
Estrogen receptor alpha
Androgen receptor
Waun Ki Hong
Genome instability
Parathyroid adenoma
MyoD
HSP90AB1
Cyclin-dependent kinase 6
Cyclin-dependent kinase 4
P16
Mitogen
TAF1
Intranodal palisaded myofibroblastoma
Endostatin
Splenic marginal zone lymphoma
George Sai Wah Tsao
HDAC3
CCNDBP1
Polyomaviridae
FBXO4
FHL2
Marginal zone B-cell lymphoma
RHOBTB2
Anti-Cyclin D1 antibody [RM241] (GTX33611) | GeneTex
Systematic validation of predicted microRNAs for cyclin D1 | BMC Cancer | Full Text
Mantle Cell Lymphoma: Practice Essentials, Overview, Pathophysiology
SUMO-modified nuclear cyclin D1 bypasses Ras-induced senescence - Ludwig Cancer Research
Expression of d1- and d3-cyclin genes in pea (Pisum sativum L.) root meristem under clinorotation
Cardiomyocyte proliferation and protection against post-myocardial infarction heart failure by cyclin D1 and Skp2 ubiquitin...
Overexpression of cyclin D1 and c-Myc gene products in human primary epithelial ovarian cancer | International Journal of...
CDK4, CDK6, Cyclin D1 and Notch1 immunocytochemical expression of oral brush liquid-based cytology for the diagnosis of oral...
The values of Transgelin, Stathmin, BCOR and Cyclin-D1 expression in differentiation between Uterine Leiomyosarcoma (ULMS) and...
Copy number variation in CCND1 gene is implicated in the pathogenesis of sporadic parathyroid carcinoma
abm | CRISPR, Viral Vectors, Cell Lines, PCR/RT/qPCR, Antibodies
C3d Positive Control Slides suitable for immunohistochemistry (formalin-fixed, paraffin-embedded sections)
Notch Signaling Pathway: R&D Systems
Recombinant Human E2F3 GST (N-Term) Protein (H00001871-P01): Novus Biologicals
Cancer Genes | CancerQuest
Novel Drug Approved for Breast Cancer: Palbociclib (Ibrance)
Blebs produced by actin-myosin contraction during apoptosis release damage-associated molecular pattern proteins before...
Frontiers | Involvement of TGF-β and Autophagy Pathways in Pathogenesis of Diabetes: A Comprehensive Review on Biological and...
ARCHIVED - Environment and Climate Change Canada - Evaluating Existing Substances - Assessment report for Benzene, 1-methyl-2...
Kencur Ginger's Anti-Cancer Effects Revealed
Resveratrol: MedlinePlus suplementos
Non-Hodgkin Lymphoma Guidelines: Classification, Diagnosis, Staging
WTS database | WHO FCTC
MMPC :: Strain
Research products - EXBIO Antibodies
Genentech: Avi Ashkenazi | Distinguished Fellow, Cancer Immunology
Gene6
- The protein encoded by this gene belongs to the highly conserved cyclin family, whose members are characterized by a dramatic periodicity in protein abundance throughout the cell cycle. (genetex.com)
- The molecular consequence of translocation is overexpression of the protein cyclin D1 (coded by the PRAD1 gene located close to the breakpoint). (medscape.com)
- Using in situ hybridization the d1- and d3-cyclin gene expression has been shown in pea ( Pisum sativum L.) root meristem cells under slow horizontal clinorotation and in the stationary conditions. (cytgen.com)
- There was no significant difference of overexpression of cyclin D1 or c-Myc gene products between these four histologic subtypes of ovarian adenocarcinomas. (bmj.com)
- It was shown that amplification of CCND1, a gene encoding cyclin D1, was more prevalent in parathyroid carcinomas than in adenomas (71 vs. 21 %, p = 0.056). (nih.gov)
- MEN 4 is caused by an inactivating mutation of the CDKN1B gene, which codes for the cyclin dependent kinase inhibitor 1B protein, also known as p27 or p27KIP1. (msdmanuals.com)
Overexpression1
- However, compared to normal ovarian tissues, overexpression of cyclin D1 and c-Myc was observed in 42.6% (20/47) and 65.9% (31/47) of tumors examined, respectively. (bmj.com)
Expression5
- Different cyclins exhibit distinct expression and degradation patterns which contribute to the temporal coordination of each mitotic event. (genetex.com)
- No abnormal expression of cyclin D1 or c-Myc was demonstrated in any of the 12 normal ovarian specimens. (bmj.com)
- Although our preliminary results need to be validated in a larger number of tumors, the abnormal expression of cyclin D1 and c-Myc in epithelial ovarian cancer reaffirms the notion that they are crucial components in the pathway of tumorigenesis and deserve further study. (bmj.com)
- Epstein-Barr virus latent membrane protein expression and cyclin DI cell cyase protein expression in malignant and normal oesophageal tissues to see whether any variation in their expression in these tissues could be of diagnostic or prognostic value. (bvsalud.org)
- EBV-LMPI protein expression and cyclin DI expression were studied immunohisto chemically in these tissue sections. (bvsalud.org)
Kinase1
- However, proliferation of cardiomyocytes induced by nuclear-targeted cyclin D1 (D1NLS) stops after one or two rounds of cell cycles due in part to accumulation of p27Kip1, an inhibitor of cyclin-dependent kinase (CDK). (elsevierpure.com)
Inhibitor1
- The new drug acts as an inhibitor of cyclin-dependent kinases 4 and 6, which are involved in promoting the growth of cancer cells, the FDA noted. (medscape.com)
Tumors1
- The second cohort was required to have tumors that had an amplification of cyclin D1, loss of p16, or both. (medscape.com)
Degradation1
- The delay of degradation of cyclin subunits is the cause of G1-phase prolongation under clinorotation leading to delay of pea seed germination. (cytgen.com)
Pathway2
- Cyclin D1 and c-Myc are key participants in the cell-cycle pathway, in which aberrancies have been associated with malignant transformation. (bmj.com)
- Thioredoxin regulates cell cycle via the ERK1/2-cyclin D1 pathway. (bvsalud.org)
Amplification1
- It was then decided that patient selection based on cyclin D amplification or p16 loss was not likely to improve patient outcome. (medscape.com)
Molecular1
- Molecular analysis of cyclin D1 modulators PRKN and FBX4 as candidate tumor suppressors in sporadic parathyroid adenomas. (cdc.gov)
Regulators2
- Cyclins function as regulators of CDK kinases. (genetex.com)
- Aims: Cyclins and other cell-cycle regulators have been used in several studies to regenerate cardiomyocytes in ischaemic heart failure. (elsevierpure.com)
Tissue1
- IHC-P analysis of human tonsil tissue using GTX33611 Cyclin D1 antibody [RM241]. (genetex.com)
Cell2
- This cyclin forms a complex with and functions as a regulatory subunit of CDK4 or CDK6, whose activity is required for cell cycle G1/S transition. (genetex.com)
- WB analysis of HeLa cell lysate using GTX33611 Cyclin D1 antibody [RM241]. (genetex.com)
CCND13
- To investigate individual susceptibility to gastroesophageal reflux disease, Barrett esophagus, and esophageal adenocarcinoma, the authors studied the frequency of the common G870A polymorphism of CCND1, which encodes cyclin D1, a key cell cycle regulatory protein. (nih.gov)
- A polymorphism (G870A) in exon 4 of the cyclin D1 (CCND1) gene creates an alternative splice site in its mRNA, encoding a protein with an altered C-terminal domain. (nih.gov)
- 8. Association between CCND1 G/A870 polymorphism, allele-specific amplification, cyclin D1 expression, and survival in esophageal and lung carcinoma. (nih.gov)
Overexpression4
- In this study, we show that overexpression of a regular component of the ERalpha transcription factor complex, cyclin D1, which occurs in approximately 40% of breast cancer patients, renders cells resistant to the new promising antiestrogen, arzoxifene. (nih.gov)
- Overexpression of cyclin D1 alters the conformation of ERalpha in the presence of arzoxifene. (nih.gov)
- No association was seen between genotype and cyclin D1 overexpression. (nih.gov)
- Overexpression of cyclin D1 is the result of bcl-1 rearrangement, a t(11;14) translocation, and is implicated in various neoplasms. (bvsalud.org)
Proliferation4
- Normal cell proliferation is closely regulated by proteins called cyclins. (ox.ac.uk)
- Hyperglycemia for 4 days of chicken embryo incubation caused a reduction in cell proliferation in cardiac tissue due to suppression of the cell cycle, specifically the upregulation of P21 and downregulation of cyclin D1. (ohsu.edu)
- Cell proliferation is tightly controlled by the cell-cycle regulatory proteins, primarily by cyclins and cyclin-dependent kinases (CDKs) in the G 1 phase. (molcells.org)
- Cyclin D1 is required for proliferation of Olig2-expressing progenitor cells in the injured cerebral cortex. (unibas.ch)
Polymorphism2
Proteins1
- Furthermore, ST water extract up-regulated expression of Cyclin D1 and CDK4 proteins. (bvsalud.org)
Oncogene2
CDK43
- 6. Cyclin D1b variant promotes cell invasiveness independent of binding to CDK4 in human bladder cancer cells. (nih.gov)
- This effect is mediated by the upregulation of a CDK inhibitor p21, which causes the decrease in cyclin D1 and CDK4 protein levels and subsequent reduction of pRb hyperphosphorylation. (molcells.org)
- Thus, the p21-cyclinD1/CDK4 signaling pathway plays a critical role in ARMS/Kidins220-dependent cell-cycle progression. (molcells.org)
Correlate2
- 0.05) but did not correlate with cyclin D1 expression. (ox.ac.uk)
- Objectives:This study was aimed to detect cyclin D1 in colorectal cancer patients and to correlate cyclin D1 expression with different pathological changes in colorectum.Methods: Tissues microarray paraffin block with 48 colorectal cancer samples were retrieved from the archives of Elrahma Medical Center. (sciencegate.app)
Correlation3
- No correlation between cyclin D1 and D2 and growth rates or CD38 expression was observed. (medscape.com)
- Cyclin D1 expression in transitional cell carcinoma of the bladder: correlation with p53, waf1, pRb and Ki67. (ox.ac.uk)
- 16. OVCA1 expression and its correlation with the expression levels of cyclin D1 and p16 in cervical cancer and intraepithelial neoplasia. (nih.gov)
Differentially2
- The D cyclins were expressed differentially in chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), and mantle cell lymphoma (MCL) with strong staining of cyclin D1 and D2 in MCL, strong staining of cyclin D1 but weak staining of cyclin D2 in 4 of 5 PLLs, and low-level staining for both cyclins in most CLLs. (medscape.com)
- 3. Expression of cyclin D1 splice variants is differentially associated with outcome in non-small cell lung cancer patients. (nih.gov)
Splice3
- 10. Alteration of cyclin D1 transcript elongation by a mutated transcription factor up-regulates the oncogenic D1b splice isoform in cancer. (nih.gov)
- 12. Cyclin D1 splice variants. (nih.gov)
- 14. Cyclin D1b splice variant promotes αvβ3-mediated adhesion and invasive migration of breast cancer cells. (nih.gov)
Alteration1
- cyclin D1 not correlates with pathological alteration of colorectal cancer. (sciencegate.app)
Progression2
- In addition, there was a significantly higher progression rate for those patients with Ta/T1 disease whose tumours demonstrated cytoplasmic cyclin D1 staining. (ox.ac.uk)
- Abstract Background: Cyclin D1 plays a vital role in cancer cell cycle progression and is overexpressed in many human cancers, including colorectal cancer. (sciencegate.app)
Immunohistochemistry2
Protein expression1
- 4. Cyclin D1b protein expression in breast cancer is independent of cyclin D1a and associated with poor disease outcome. (nih.gov)
Prognostic3
- The results indicate that high-resolution flow cytometric evaluation of the D cyclins discriminates between the CD5+ B-cell lymphoproliferative disorders and might impart prognostic information in CLL. (medscape.com)
- In a multivariate analysis, cyclin D1 expression is an independent prognostic indicator of survival in those patients with muscle-invasive disease. (ox.ac.uk)
- 1. Expression Of Cyclin D1 Protein Isoforms And Its Prognostic Significance In Cervical Cancer. (nih.gov)
Significantly2
- However, cyclin D1 levels were significantly higher in ZAP-70+ CLL cases, although no association between ZAP-70 and cyclin D2 was detected. (medscape.com)
- These results indicate that cyclin D1 expression is significantly higher in low-stage, well differentiated bladder tumours and strongly correlates with waf1 expression. (ox.ac.uk)
Decrease1
- Following siRNA knockdown of Raf-1, Western blot analysis revealed a significant decrease in Raf-1, cyclin D1, and all p-4EBP1 forms noted above. (nih.gov)
Outcome1
- 2. Cyclin D1b in human breast carcinoma and coexpression with cyclin D1a is associated with poor outcome. (nih.gov)
Growth1
- 9. Cyclin D1b variant influences prostate cancer growth through aberrant androgen receptor regulation. (nih.gov)
Patients1
- L'étude portait sur 20 patients atteints d'un carcinome gastrique, 20 patients souffrant d'une gastrite à H. pylori et 20 personnes en tant que groupe témoin. (who.int)
Lack1
- 15. Enhanced skin carcinogenesis and lack of thymus hyperplasia in transgenic mice expressing human cyclin D1b (CCND1b). (nih.gov)
Cells1
- The impact of Cyclin D1 on proliferating cells in the intact and injured mouse cortex. (unibas.ch)