scispace - formally typeset
Search or ask a question

Is foxo4 upregulated in senescent cells? 


Best insight from top research papers

Yes, FOXO4 is indeed upregulated in senescent cells. Studies have shown that FOXO4 is progressively upregulated after senescence induction and is found in damage foci in senescent cells, where it inhibits apoptosis through interacting with p53 . Additionally, FOXO4 has been found to be specifically expressed in human Leydig cells and translocates to the nucleus in the elderly, contributing to decreased testosterone synthesis. By disrupting the FOXO4-p53 interaction, a specific FOXO4 blocker, FOXO4-DRI, selectively induces p53 nuclear exclusion and apoptosis in senescent Leydig cells, highlighting the role of FOXO4 in maintaining the viability of senescent cells . These findings collectively support the upregulation and functional significance of FOXO4 in senescent cells.

Answers from top 4 papers

More filters
Papers (4)Insight
FOXO4-DRI selectively removes senescent cells but does not upregulate FOXO4. It targets senescent chondrocytes by disrupting FOXO4-p53 interaction, inducing apoptosis, and enhancing cartilage quality.
Yes, FOXO4-DRI targets myofibroblasts in pulmonary fibrosis by decreasing senescent cells and downregulating the senescence-associated secretory phenotype, indicating FOXO4 involvement in senescent cell regulation.
Yes, FOXO4 is progressively upregulated in senescent cells, where it inhibits apoptosis by interacting with p53, as highlighted in the research paper.
FOXO4 interacts with p53 to induce cellular senescence by upregulating p21, but the paper does not specify if FOXO4 itself is upregulated in senescent cells.

Related Questions

Can foxo1 binds to NLRP3 promoter region?5 answersYes, FoxO1 can bind to specific promoter regions, but the target varies depending on the context. In the provided research, FoxO1 was found to interact with the NLRP3 inflammasome signaling in osteoblasts under oxidative stress. Additionally, FoxO3 was identified to bind to the NOX4 promoter region in the context of neuronal ferroptosis after intracerebral hemorrhage. Furthermore, FoxO1 was shown to inhibit STAT3-mediated leptin signaling by directly interacting with STAT3, affecting the POMC promoter complex. These studies highlight the diverse roles of FoxO proteins in binding to different promoter regions to regulate various cellular processes, emphasizing the versatility and importance of these transcription factors in mediating gene expression and cellular responses.
What are the biomarkers of cell senescent in cancer?5 answersCell senescence in cancer can be identified by specific biomarkers such as senescence-associated β-galactosidase (SA-β-gal), cell division cycle 7-related protein kinase, partitioning defective three homolog B, putative ATP-dependent RNA helicase DHX57. These biomarkers play a crucial role in detecting therapy-induced senescence (TIS) in cancer cells, aiding in prognosis and treatment strategies. Studies have shown that the expression of markers like p21CIP1, H3K9Me3, and Lamin B1 can indicate a senescence-like phenotype in breast cancer samples post neoadjuvant chemotherapy, with Lamin B1 showing promise as a predictive marker for senescence detection. The identification of these biomarkers is essential for understanding the impact of senescence on cancer progression and developing targeted therapies for age-related diseases like cancer.
Can senescent cells show cell proliferation?5 answersSenescent cells can show cell proliferation, as indicated by the presence of the proliferation marker Ki-67 in the tumor invasion front of prostate cancer cells. Additionally, senescent cells have been shown to stimulate skin carcinogenesis by elevating p38MAPK and MAPK/ERK signaling. However, the replicative capacity of senescent endothelial cells (ECs) can be improved by cultivation on micro-structured surfaces with anisotropic topography, leading to enhanced cell proliferation. The senescence-associated secretory phenotype (SASP) secreted by senescent cells can have pro-inflammatory and growth-stimulatory effects, contributing to different stages of carcinogenesis. In a study using HeLa cells, the morphology of senescent cells varied depending on the culture platform, with flattened morphology observed in 2D culture systems but reduced artifact in 3D scaffolds, suggesting that the 3D scaffolds suppress senescence in cells.
Can mRNA therapy be used to clear senescent cells?5 answersmRNA therapy has the potential to be used to clear senescent cells, which are implicated in age-related declines and diseases. Senescent cells accumulate in tissues with advancing age and can have detrimental effects. Understanding the molecular mechanisms by which MicroRNAs influence senescence can enable diagnostic and therapeutic opportunities directed at senescent cells. Additionally, recent progress has been made in the field of senescent cell clearance and various anti-aging strategies, including the targeted killing of senescent cells. Removing senescent cells from the body has been shown to delay the occurrence of aging-associated diseases. While the abstracts do not explicitly mention mRNA therapy for clearing senescent cells, the potential of mRNA therapeutics in clinical applications is highlighted, indicating that mRNA therapy could be explored as a potential approach for clearing senescent cells.
What is role of transcriptional factor FOXO1 in progression of gestational diabetes?4 answersThe transcription factor FOXO1 plays a role in the progression of gestational diabetes. It is involved in regulating cellular processes such as apoptosis, autophagy, oxidative stress, and metabolic dysregulation. FOXO1's function depends on the modulation of downstream targets, including apoptosis- and autophagy-associated genes, anti-oxidative stress enzymes, cell cycle arrest genes, and metabolic and immune regulators. In pregnant women with gestational diabetes mellitus (GDM), FOXO1 expression is higher in the placenta and omental adipose tissue compared to those with normal glucose tolerance. FOXO1 has been found to have a potential role as a pro-inflammatory factor in GDM and may be involved in the development of insulin resistance through interaction with tumor necrosis factor-α (TNF-α). These findings suggest that FOXO1 may contribute to the pathogenesis of gestational diabetes by affecting various cellular processes and interacting with inflammatory factors.
What is role of transcriptional factor FOXO1 in progression of gdm?5 answersThe transcription factor FOXO1 plays a role in the progression of gestational diabetes mellitus (GDM). FOXO1 is involved in various diseases, including cardiovascular diseases, diabetes, and cancer. In the context of GDM, FOXO1 is implicated in cardiac hypertrophy, which can lead to heart failure and cardiovascular events. FOXO1 regulates cardiac growth/protein synthesis, calcium homeostasis, cell apoptosis, and autophagy, and is controlled by upstream signaling molecules such as phosphatidylinositol 3-kinase/Akt and AMP-activated protein kinase. It also regulates downstream transcription proteins like ubiquitin ligases muscle RING finger 1/muscle atrophy F-box and calcineurin/nuclear factor of activated T cells. Targeted regulation of FOXO1 is considered an effective method of intervening in myocardial hypertrophy, suggesting its potential role in the treatment of cardiac hypertrophy associated with GDM.

See what other people are reading

How to make people younger by cell therapy ?
5 answers
Cell therapy can potentially reverse aging by utilizing innovative techniques. Anti-Aging Regenerative Cosmetology (AARC) combines myoblasts and foreskin fibroblasts to enhance appearance and function, harnessing their regenerative properties for genetic cell therapy. Stem cells, derived from personalized sources, can regenerate organs like the heart, providing a youthful replacement for damaged tissues. Multipotent adult cells offer a novel approach to treat aging and diseases through transplantation to damaged areas. These advancements hold promise for sustaining good health, appearance, and life prolongation by improving cell genetics and organ functions. By leveraging the regenerative potential of various cell types, cell therapy emerges as a cutting-edge solution to combat aging and enhance overall well-being.
What use does the cmv promoter have in viral vector vaccines?
4 answers
The CMV promoter plays a crucial role in viral vector vaccines by driving the transcription of recombinant genes in host cells, such as HEK293 cells. This promoter is utilized in cytomegalovirus (CMV) vectors, which have been explored as vaccine vectors for various pathogens, including influenza and coronaviruses. CMV vectors expressing antigenic peptides or proteins can induce robust immune responses, leading to long-term cellular immunity and humoral responses against respiratory pathogens. The use of CMV as a vaccine vector has shown promising results in generating protective immunity against infectious diseases in animal models, highlighting its potential for human vaccine development. Overall, the CMV promoter in viral vector vaccines is essential for efficient gene expression and the induction of protective immune responses against a range of pathogens.
How does the disruption of circadian rhythms affect arthritis?
4 answers
Disruption of circadian rhythms has been shown to significantly impact arthritis by affecting joint health and inflammatory responses. Studies have highlighted that chronic circadian rhythm disturbance can induce osteoarthritis-like degeneration in cartilage, accelerating matrix degradation and inflammation. Furthermore, alterations in the circadian clock within articular cartilage have been linked to a procatabolic state, exacerbating the pathogenesis of diseases like osteoarthritis. Research also suggests that circadian rhythm disturbances can lead to increased symptom severity in rheumatoid arthritis (RA) patients, affecting their quality of life and potentially worsening inflammation and pain levels. These findings underscore the critical role of circadian rhythms in arthritis pathophysiology and highlight the potential therapeutic implications of chronotherapy in managing arthritis conditions.
What activates myfibroblasten?
5 answers
Myofibroblasts are activated by various stimuli, leading to their secretory and contractile phenotype. Activation traditionally involved the de novo expression of alpha-smooth muscle actin stress fibers, previously viewed as a binary process. Recent studies have shifted towards considering myofibroblast activation as a continuous spectrum, necessitating a method to quantify this activation level. Mechanisms of myofibroblast activation include responses to changes in the cytokine milieu and extracellular matrix composition, leading to dynamic phenotypic alterations. Additionally, compounds like cyclolanostane and rophenol have been identified as fibroblast activators, promoting the production of collagen and other extracellular matrix components. These findings collectively highlight the multifaceted nature of myofibroblast activation and the diverse pathways involved in their stimulation.
What are the potential therapeutic targets for modulating myofibroblast activation in fibrosis?
5 answers
Potential therapeutic targets for modulating myofibroblast activation in fibrosis include targeting the Rho/myocardin-related transcription factor (MRTF) / serum response factor (SRF) pathway, TGF-β/Smad, MAPK/P38/ERK/JNK, PI3K/AKT, and WNT/β-catenin signaling pathways regulated by non-coding RNAs, galectin-3, PTX2, and PTX3/CD44 axis inhibitors, as well as MRC2, IGFBP7, and NT5DC2 identified through proteomic analysis. These targets aim to block myofibroblast activation, collagen synthesis, and pro-fibrotic mediator production, ultimately reducing fibrosis progression. Additionally, exploring next-generation epigenetic regulatory targets may revolutionize treatment strategies for cardiac fibrosis associated with cardiovascular diseases. Targeting these pathways and molecules presents promising avenues for developing effective anti-fibrotic therapies to combat fibrosis-related diseases.
Are promoters and enhancers characterised by accessible chromatin?
4 answers
Promoters and enhancers are indeed characterized by accessible chromatin. Accessible chromatin, indicative of active gene regulatory regions, commonly harbors enhancers and promoters. Studies across various species, including humans, mice, pigs, cattle, and bumblebees, have utilized techniques like ATAC-seq to identify and characterize regulatory elements. These elements are often tissue-specific, enriched with binding motifs of specific transcription factors, and crucial for gene expression regulation. The conservation of regulatory elements between species varies, with promoters showing more conservation than distal enhancers. Understanding the chromatin accessibility at promoters of key host factor genes like ACE2 and TMPRSS2 has provided insights into the tropism mechanisms of viruses like SARS-CoV-2, emphasizing the pivotal role of accessible chromatin in gene regulation and disease pathogenesis.
Are promoters and enhancers in humans characterised by accessible chromatin?
4 answers
Promoters and enhancers in humans are indeed characterized by accessible chromatin. Accessible chromatin is strongly associated with active gene regulatory regions, including enhancers and promoters, which are crucial for gene expression control. Studies have shown that the chromatin accessibility landscape differs between inner cell mass (ICM) and trophectoderm (TE) in human preimplantation embryos, highlighting the epigenetic regulation of cell lineage specification during early embryo development. Additionally, genome-wide profiling of chromatin accessibility in Plasmodium falciparum revealed that chromatin accessibility by ATAC-seq is predictive of active transcription and histone marks, aiding in the identification of regulatory regions such as promoters and enhancer-like elements. These findings underscore the importance of accessible chromatin in characterizing promoters and enhancers across different biological contexts in humans.
Ampk activation in L6 cells via mitochondria
4 answers
AMPK activation in L6 cells can be linked to mitochondria through various mechanisms. AMPK activation, induced by compounds like AICAR and caffeine, has been shown to increase mitochondrial biogenesis in L6 myotubes. Additionally, acetic acid has been found to enhance glucose uptake and fatty acid metabolism in skeletal muscle cells through AMPK activation, leading to increased expression of mitochondrial markers like GLUT4 and myoglobin. Furthermore, flavonoids from Psidium guajava leaves have demonstrated significant AMPK activation in L6 cells, suggesting a potential role in regulating mitochondrial function. Overall, AMPK activation in L6 cells appears to play a crucial role in promoting mitochondrial biogenesis and metabolic adaptations, highlighting the intricate connection between AMPK signaling and mitochondrial function in these cells.
Are there papers that show actin cytoskeleton in chondrocytes for osteoarthritis?
5 answers
Studies have highlighted the significance of the actin cytoskeleton in chondrocytes for osteoarthritis (OA) progression. Research has shown that regulation of actin cytoskeleton plays a crucial role in OA development. Furthermore, investigations have demonstrated that autophagy and cytoskeleton integrity are major factors in OA progression, with nesfatin-1 being linked to the suppression of excessive autophagy and improvement of cytoskeleton integrity in OA chondrocytes. Additionally, the actin cytoskeleton is involved in controlling chondrocyte phenotype through processes like actin polymerization and stress fiber formation, which are regulated by RhoA/ROCK signaling pathways. These findings collectively emphasize the importance of understanding and targeting the actin cytoskeleton in chondrocytes for better management and potential therapeutic interventions in osteoarthritis.
How does the UPR of mitochondria recognize unfolded proteins?
5 answers
The recognition of unfolded proteins by the mitochondrial unfolded protein response (UPRmt) involves the integration of signals from mitochondrial misfolding stress (MMS) into the nucleus. The UPRmt is activated by the release of two key signals: cytosolic-mitochondrial reactive oxygen species (mtROS) and the accumulation of mitochondrial protein precursors in the cytosol (c-mtProt). Mitochondrial stress leads to the release of mtROS into the cytosol and causes defects in mitochondrial protein import, resulting in c-mtProt accumulation. These signals converge to activate the UPRmt pathway, where mtROS oxidize cytosolic HSP40 protein DNAJA1, enhancing the recruitment of cytosolic HSP70 to c-mtProt. This ultimately leads to the translocation of HSF1 to the nucleus, initiating the transcription of UPRmt genes.
How do plants sense phosphate?
5 answers
Plants sense phosphate levels primarily through inositol polyphosphates (InsPs) interacting with SPX domain-containing proteins, such as SPX1. These interactions lead to the inhibition of PHR transcription factors, crucial for maintaining phosphate homeostasis in plants. Additionally, recent research highlights the role of inositol pyrophosphate InsP 8 in regulating the activity of PHR through its interaction with SPX domain proteins. The SPX-InsP signaling module is evolutionarily conserved and plays a vital role in plant Pi transport and signaling systems. Furthermore, advancements in the development of genetically encoded fluorescent sensors, like FLIPPi, enable real-time monitoring of cellular phosphate concentrations in plants, aiding in understanding the molecular mechanisms of Pi transport and homeostasis.