scispace - formally typeset
Search or ask a question

What role do microRNAs play in the regulation of apoptosis in cancer cells? 


Best insight from top research papers

MicroRNAs (miRNAs) play a crucial role in regulating apoptosis in cancer cells. They can act as either apoptotic or anti-apoptotic factors by modulating the expression levels of tumor inhibitor or oncogene genes, thus impacting the apoptosis pathways in cancer. Specifically, miRNAs like miR-192, miR-194, miR-215, miR-22, and miR-101 have been identified to be involved in apoptosis regulation in cancer cells. These miRNAs can influence key factors in the apoptotic pathways, such as p53 and its downstream targets like p21 and Mdm2, ultimately affecting cell survival and proliferation. Understanding the mechanisms by which miRNAs regulate apoptosis in cancer cells provides valuable insights for developing novel therapeutic strategies targeting these miRNAs to modulate apoptosis and improve cancer treatment outcomes.

Answers from top 4 papers

More filters
Papers (4)Insight
MicroRNAs regulate apoptosis in cancer cells, impacting cell survival. They act as post-transcriptional gene expression regulators, influencing pathways crucial for cancer development and progression.
MicroRNAs like miR-192/194/215 regulate apoptosis in cancer cells by influencing p53 activity, inducing cell cycle arrest, and modulating p53 regulators like Mdm2 and Chk2.
MicroRNAs can act as apoptotic or anti-apoptotic factors in cancer cells by modulating tumor inhibitor or oncogene gene expression, influencing apoptosis regulation in prostate cancer.
MicroRNAs regulate apoptosis in cancer cells by acting as oncogenes or tumor suppressors, influencing gene expression, and serving as potential biomarkers for diagnosis and prognosis.

Related Questions

How does the dysregulation of microRNA expression affect apoptosis in different types of cancer?5 answersDysregulation of microRNA (miRNA) expression significantly impacts apoptosis in various cancers. MiRNAs play crucial roles in regulating cell death pathways. In colorectal cancer (CRC), dysregulated miRNAs influence the expression of genes involved in apoptosis, such as BIRC5, CTSS, and CSF2R, potentially affecting CRC-specific survival. Furthermore, miRNAs act as tumor suppressors or oncogenes by modulating apoptotic pathways through gene silencing. In skin cancer induced by UVB radiation, altered miRNA expression levels in tumor tissues compared to normal skin suggest their involvement in photocarcinogenesis, highlighting their potential as prognostic biomarkers and therapeutic targets. Understanding the intricate relationship between dysregulated miRNAs and apoptosis is crucial for developing targeted cancer therapies.
What is the role of microRNAs in the communication of tumor microenvironment?4 answersMicroRNAs (miRNAs) play a crucial role in the communication between tumor cells and components of the tumor microenvironment (TME). They are small noncoding RNAs that regulate gene expression at the post-transcriptional level. In the TME, miRNAs are involved in the complex interactions between tumor cells and various cell types, including endothelial cells, fibroblasts, immune cells, and signaling molecules. miRNAs have been shown to be central communicators between the immune system and cancer, and they are secreted in high copy numbers from cancer cells. Exosomes containing miRNAs have been found to play a crucial role in the crosstalk between tumor cells and cancer-associated fibroblasts (CAFs), promoting tumor progression. miRNAs also modulate the behavior of immune cells in the TME, affecting tumor growth and progression. Understanding the role of miRNAs in the communication of the TME can help in the development of targeted therapies and diagnostic approaches for cancer treatment.
What is the The role of microRNA in Triple-Negative Breast Cancer?5 answersMicroRNAs (miRNAs) play a crucial role in triple-negative breast cancer (TNBC) by affecting its occurrence, invasion, metastasis, prognosis, and response to chemotherapy. Certain miRNAs have been identified as potential biomarkers for TNBC diagnosis and prognosis. miR-22, for example, is an oncogenic miRNA that promotes epithelial-mesenchymal transition (EMT), tumor invasion, and metastasis in hormone-responsive breast cancer, as well as in TNBC. Inhibition of miR-22 has shown promise in reducing EMT and suppressing metastatic spread in TNBC. Other miRNAs, such as miR-155, miR-182-5p, miR-9-1-5p, miR-200b, and miR-22-3p, have been implicated in TNBC diagnosis and therapy. Additionally, miRNAs have been associated with DNA repair deficiency and immune cell infiltration in TNBC, further highlighting their importance in the disease. Overall, miRNAs have emerged as potential therapeutic targets and prognostic markers for TNBC.
What are the effects of miRNAs on gene expression?4 answersMicroRNAs (miRNAs) are small non-coding RNA molecules that regulate gene expression at the posttranscriptional level. They can bind to complementary sequences on cellular mRNAs, leading to mRNA degradation or inhibition of translation. Plant-derived miRNAs have been shown to regulate gene expression in mammals through cross-kingdom regulation. In addition, miRNAs play a role in stabilizing the transcriptional regulatory network by attenuating the degradation of target genes and maintaining transcriptional homeostasis. They can reduce cell-to-cell variability in protein expression and induce correlations between target expression levels. Furthermore, miRNAs can act as "competing RNAs" and influence the expression of other targets. Overall, miRNAs have a significant impact on gene expression by regulating mRNA stability, translation, and transcriptional dynamics.
What is the role of microRNA in hemedynamics?5 answersMicroRNAs play a crucial role in hematodynamics, including normal hematopoiesis and hematopoietic malignancies. They are involved in the differentiation of B and T lymphocytes, erythroid differentiation, hematopoietic differentiation, and myeloid differentiation. Aberrant expression of microRNAs has been associated with solid tumors and hematopoietic malignancies, such as chronic lymphocytic leukemia, diffuse large B-cell lymphomas, and acute myeloid leukemia. In the liver, microRNAs are also involved in ischemia-reperfusion (I/R) injuries, which can occur during hepatic surgery or transplantation. MicroRNAs regulate the expression of cellular participants and humoral factors associated with I/R injury, and they have the potential to be used as biomarkers and pharmacological targets for the prevention, diagnosis, and treatment of I/R injuries. Additionally, microRNAs have been shown to regulate gene expression and play a role in somite segmentation during early vertebrate development. Overall, microRNAs have emerged as important players in hematodynamics and have potential therapeutic applications in various diseases, including cancer.
What are the most interesting microRNAs in cancer biology research?5 answersMicroRNAs (miRNAs) have been extensively studied in cancer biology research. Several miRNAs have been identified as interesting targets in cancer research. For example, miR-18a, miR-21, miR-143/145, miR-210, miR-218, and miR-221 have shown dysregulation in multiple cancer types and have potential as biomarkers for diagnosis and prognosis. These miRNAs have been found to play roles in various aspects of cancer, including tumor initiation, drug resistance, and metastasis. Additionally, miRNAs have been implicated in the regulation of tumor metabolism, with altered expression patterns of tumor suppressor miRNAs being frequently observed in cancers. Furthermore, miRNAs have been shown to be involved in the immune modulation, vascularization, and altered stress response, which are important cancer hallmarks. Overall, miRNAs have emerged as promising targets for cancer research, with potential applications as diagnostic markers and therapeutic targets.

See what other people are reading

Do mushrooms have anticancer properties?
5 answers
Mushrooms exhibit significant anticancer properties due to their bioactive compounds. Research has highlighted that mushrooms contain molecules with therapeutic potential, such as β-glucans, ergosterol, and various acids, which can inhibit cancer cell proliferation, prevent carcinogenesis, and hinder metastasis. These bioactive constituents interfere with crucial proteins involved in cancer progression, like epidermal growth factor receptor and vascular endothelial growth factor, thereby impeding tumor growth and metastasis. Additionally, mushrooms possess immunomodulatory abilities, anti-angiogenic properties, and can enhance chemosensitivity, making them promising candidates for cancer management. The diverse anticancer activities of mushroom polysaccharides and triterpenoids have been extensively studied, showcasing their potential in preventing and treating cancer through mechanisms like apoptosis induction and immune response regulation.
How a guta6thione precursor impact infant cancer?
4 answers
A gut microbiota-derived short-chain fatty acid, propionate, plays a crucial role in influencing the development of diseases, including cancer, in infants. Maternal obesity, which can impact the gut microbiota composition of offspring, has been linked to an increased risk of aggressive and treatment-resistant breast cancer in daughters, potentially affecting their response to cancer therapies. Additionally, the gut virome composition, influenced by various environmental factors like having older siblings and urban living, can impact the metabolic capacity of bacterial hosts and predispose infants to autoimmune and metabolic disorders. Furthermore, human milk oligosaccharides (HMOs) like 2'-fucosyllactose (2'-FL) in infant formula can support the production of secondary microbial metabolites, such as bile acids, at levels comparable to breastfed infants, potentially affecting systemic metabolism and gut microbiome function. These factors collectively highlight the intricate relationship between gut microbiota, maternal influences, and infant health outcomes, including cancer susceptibility.
How glutathione precursor cysteine impact in child cancer?
5 answers
Cysteine, a precursor of glutathione (GSH), plays a crucial role in pediatric cancer. Cysteine's unique nucleophilicity allows it to participate in metabolic pathways, redox homeostasis, and protein modifications, influencing cellular states and functions. GSH, derived from cysteine, acts as a major intracellular antioxidant, impacting cell differentiation, proliferation, and apoptosis, essential processes in cancer development. Additionally, glutathione S-transferases (GSTs), which utilize GSH, are implicated in detoxification and disease susceptibility, including pediatric cancers, offering opportunities for therapeutic exploitation. Disturbances in GSH homeostasis, regulated by cysteine availability, are linked to tumor initiation, progression, and treatment response, with elevated GSH levels promoting resistance to chemotherapy in cancer cells. Understanding the interplay between cysteine, GSH, and pediatric cancer biology is crucial for developing effective therapeutic strategies.
What is the influence of Nanjing Line 1?
5 answers
The influence of Nanjing Line 1, as studied in various research papers, shows a significant positive impact on property values within its proximity. Both theoretical and empirical analyses confirm that properties within 500 meters of Line 1 experience a 14.3% increase in value compared to those farther away, with values remaining higher up to 1,500 meters. This effect diminishes as distance increases, with the most substantial influence observed at around 320 meters from the subway station. Additionally, the impact of urban rail transit, like Line 1, on property values exhibits a sub-market effect, with rural markets experiencing a greater influence than urban markets.
What is effects of tetracyclines, macrolides, quinolones and sulfonamides in fish (danio rerio and Daphnia magna)?
4 answers
Tetracyclines, macrolides, quinolones, and sulfonamides, such as oxytetracycline (OTC), florfenicol (FF), and sulfamethoxazole (SMX), have varying effects on fish like zebrafish (Danio rerio). OTC and FF exposure in zebrafish larvae led to increased movement and exploration, reduced anxiety-like behavior, and compromised optomotor responses. SMX exposure caused severe histopathological liver damage, altered gene expression related to fatty acid metabolism, and dysbiosis in gut microbiota. Additionally, zebrafish exposed to SMX and clarithromycin (CLA) exhibited higher bacterial loads, altered immune mechanisms, and decreased immune response to viral infection, indicating immunosuppression. These findings highlight the diverse impacts of these antibiotics on fish physiology, behavior, immune responses, and microbiota, emphasizing the need for comprehensive assessment of antibiotic effects in aquatic organisms.
What is known about tyrosine kinase inhibitors and chlamydia infections?
5 answers
Tyrosine kinase inhibitors have been implicated in modulating Chlamydia infections. Research suggests that Chlamydia species associated with human infections, such as C. trachomatis and C. pneumoniae, exhibit unique requirements for Src-family kinases during their developmental cycle, involving tyrosine phosphorylation and microtubule-dependent trafficking. Additionally, the JAK-STAT3 signaling pathway plays a role in the anti-apoptotic effect of C. psittaci infection, where tyrosine kinase inhibitor AG-490 increased apoptosis in infected cells. Furthermore, the epidermal growth factor receptor (EGFR) and transforming growth factor beta (TGF-β) signaling pathways cooperate during Chlamydia infection, with inhibition of both pathways significantly reducing inclusion development and preventing epithelial-mesenchyme transition (EMT) induction. These findings highlight the potential of tyrosine kinase inhibitors as targets for treating Chlamydia infections.
Isoforms of the protein clusterin
4 answers
Clusterin (CLU) has multiple isoforms with distinct functions. In Alzheimer's disease, a non-glycosylated 45 kDa isoform called mitoCLU is found in the mitochondrial matrix of neurons and astrocytes, contributing to disease development. In osteoarthritis, two isoforms of clusterin have opposing roles: secreted clusterin (sCLU) acts as a cytoprotective chaperone, while nuclear clusterin (nCLU) promotes cell death. Additionally, CLU isoforms play a role in oral squamous cell carcinoma (OSCC), where abnormal expression is linked to cancer progression, but the exact roles in carcinogenesis remain unclear. The presence of sCLU and nCLU isoforms has also been observed in a primary culture of porcine aortic endothelial cells, with sCLU being the predominant form influenced by lipopolysaccharide exposure.
What are the mechanisms behind repression of pancreatic beta cell disallowed genes?
5 answers
The repression of disallowed genes in pancreatic beta cells involves intricate mechanisms. Epigenetic regulation by Polycomb Repressive Complexes (PRC) plays a crucial role. Histone modifications and DNA methylation, particularly via DNMT3A, contribute to the repression of disallowed genes during beta cell maturation. Additionally, microRNAs (miRNAs) are implicated in gene disallowance, with miRNA depletion leading to upregulation of specific disallowed genes like Fcgrt, Oat, and Pdgfra. Furthermore, the selective repression of certain genes involved in cellular proliferation, such as Yap1 and Igfbp4, is observed in beta cells, indicating a mechanism to control beta cell growth and insulin production. These findings highlight the complex interplay of epigenetic, transcriptional, and posttranscriptional regulatory mechanisms in maintaining the unique gene expression profile of pancreatic beta cells.
Is KLK7 assoacited with cancer?
5 answers
KLK7, a kallikrein-related peptidase, is indeed associated with cancer. Research indicates that KLK7 is overexpressed in various tumor types, including squamous cell carcinomas, ovarian and endometrium cancers, salivary gland tumors, and adenocarcinomas of the upper gastrointestinal tract. In breast and lung cancers, the long non-coding RNA KRT7-AS acts as a tumor suppressor by regulating KLK7 levels, impacting tumorigenesis and apoptosis. Moreover, in basal-like breast cancer, KLK7 mRNA levels are significantly higher, potentially influencing patient survival outcomes. Additionally, in ovarian cancer, elevated KLK7 expression is associated with disease progression and poorer prognosis. Notably, in triple-negative breast cancer, high KLK7 protein levels are linked to improved overall survival, suggesting a favorable prognostic value in this specific cancer subtype.
How is MIF involved in angiogenesis of prostate cancer?
5 answers
Macrophage migration inhibitory factor (MIF) plays a crucial role in angiogenesis in prostate cancer. MIF, a proinflammatory cytokine, is released by stromal and tumor cells, promoting malignant growth and metastasis by stimulating tumor cell proliferation, inhibiting apoptotic death, facilitating extracellular matrix invasion, and enhancing angiogenesis. Studies suggest that MIF, acting through its receptor CD74, is a pro-tumorigenic factor in genitourinary cancers, including prostate cancer, by promoting angiogenesis and other cancer hallmarks. Additionally, MIF inhibition in preclinical models reduces angiogenesis and tumor aggressiveness by downregulating key signaling pathways, such as ERK and AKT. Understanding the role of MIF in angiogenesis is crucial for developing novel therapeutic strategies targeting this pathway in prostate cancer.
What's MIF's role in angiogenesis of prostate cancer?
5 answers
Macrophage migration inhibitory factor (MIF) plays a crucial role in angiogenesis in prostate cancer. MIF is known to enhance malignant growth and metastasis by stimulating tumor cell proliferation, suppressing apoptotic death, facilitating invasion of the extracellular matrix, and promoting angiogenesis. Studies suggest that inhibiting MIF activity reduces the phenotypic hallmarks of cancer, including angiogenesis, by downregulating signaling pathways like ERK and AKT. Additionally, MIF can promote tumorigenesis by acting directly on cancer cells and reversing immunosuppression. Targeting oxidized MIF (oxMIF), a disease-related isoform, has shown promising results in reducing cancer cell invasion, angiogenesis, and proinflammatory cytokine production. Furthermore, the Nodal/ALK4 pathway, regulated by miR-185, is crucial in prostate cancer angiogenesis, highlighting a potential therapeutic target to inhibit angiogenesis and tumor progression.