scispace - formally typeset
Search or ask a question

What is known about beta cell gene expression and maturation postnatally. until what age are beta cells considered mature? 


Best insight from top research papers

Postnatally, beta cell maturation is a crucial process for establishing functional maturity. Key genes associated with human pancreatic beta cell maturation include Urocortin-3 (UCN3), CHGB, G6PC2, FAM159B, GLUT1, IAPP, and ENTPD3, which show developmental correlation with functional maturation. The transition from immature to mature beta cells involves the initiation of functional maturation before birth, with completion in early postnatal life. Glucose-induced insulin secretion, a hallmark of mature beta cells, is achieved postnatally after a phase of intense proliferation, crucial for establishing an appropriate functional beta cell mass. The neonatal period is decisive for acquiring an adequate functional beta cell mass, with glucose-responsive insulin secretion being a key indicator of maturity. The exact age at which beta cells are considered fully mature is not explicitly mentioned in the provided contexts.

Answers from top 5 papers

More filters
Papers (5)Insight
Postnatally, beta cell gene expression undergoes maturation, with intense proliferation before glucose-induced insulin secretion. Key regulator Scrt1 controls beta cell proliferation, and maturity extends beyond the neonatal period.
Open accessJournal ArticleDOI
Tom Barsby, Timo Otonkoski 
04 Mar 2022-Diabetologia
14 Citations
Postnatally, beta cell maturation involves shifts in energy-sensing pathways and nutrient exposure, leading to functional maturity. Beta cells are considered mature post-weaning and continue to develop thereafter.
Postnatal beta-cell gene expression transitions from cell development pathways to metabolic processes. Beta cells are considered mature around 60 days postnatal age.
Beta cell maturation markers like CHGB, FAM159B, G6PC2, GLUT1, IAPP, and ENTPD3 show increased expression postnatally, indicating maturity onset after birth. Age of beta cell maturity not specified.
Postnatal beta cells undergo maturation marked by upregulation of NEUROD1, UCN3, ABCC8, and CASR genes. Beta cells are considered mature after achieving full glucose responsiveness postnatally.

Related Questions

What gene expression is regulated by F-actin in beta cell?5 answersF-actin plays a crucial role in regulating gene expression in beta cells. Studies have shown that genes such as serum amyloid A3 (Saa3) and CXC-chemokine ligand 5 (Cxcl5) are involved in cytoskeletal rearrangement and are regulated by F-actin through the signaling pathways mediated by β-catenin. Additionally, nuclear F-actin enhances the nuclear accumulation and transcriptional function of β-catenin, influencing downstream target genes like c-myc, cyclin D, and OCT4 in the Wnt/β-catenin signaling pathway. Furthermore, APPL2, through its interaction with Rac1, promotes F-actin remodeling, affecting genes related to glucose-stimulated insulin secretion (GSIS) in pancreatic beta cells. Overall, F-actin's regulation of gene expression in beta cells is crucial for processes like insulin secretion and cell differentiation.
When do beta cells start expanding in mice?4 answersBeta cells start expanding in mice during pregnancy, specifically at gestational day 9. In female C57Bl/6 mice, the proportion and fraction of insulin-expressing but glucose-transporter-2-low (Ins+Glut2LO) progenitor cells, which are present in extra-islet β-cell clusters, undergo proliferation at gestational day 9, preceding the increase in β-cell mass and proliferation. This proliferation of Ins+Glut2LO cells within clusters continues until gestational day 15. Additionally, the overall number of clusters increases significantly at gestational day 9. This suggests that Ins+Glut2LO cells contribute to the expansion of β-cell mass during pregnancy.
What are the factors that influence maturation?5 answersFactors that influence maturation include environmental cues such as photoperiod and temperature. Other factors include social environment, mate competition, and adult sex ratio (ASR). Intense sexual selection on males, indicated by polygamous mating and male-skewed sexual size dimorphism, correlates with delayed maturation in males. In species with a female-skewed ASR, males experience later maturation. Additionally, within-group contest competition and group size can also affect maturation age in females. Undernutrition, obesity, ethnic/racial background, social class, familial characteristics, climate, and altitude are environmental factors that can influence the level of maturity at a given point in time. These factors collectively influence the evolution of maturation, a major life history trait.
At what age does the prefrontal cortex becomes fully mature?5 answers
How long does it take for neurons to mature?7 answers
When do B cells become mature?6 answers

See what other people are reading

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.
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.
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.
What is the mir-150 function?
5 answers
MicroRNA-150 (miR-150) plays diverse roles in various physiological and pathological conditions. In the context of immune-related diseases, miR-150 is crucial in regulating B cell function, immune homeostasis, and disease progression, serving as a diagnostic biomarker and prognostic tool. In heart failure, miR-150 contributes to cardioprotection by inhibiting maladaptive remodeling post-myocardial infarction and regulating unique noncoding RNA and gene signatures in cardiomyocytes. Additionally, miR-150's interaction with long noncoding RNA MIAT and downstream target Hoxa4 is pivotal in ischemic heart failure, highlighting a novel regulatory mechanism in cardiac protection. Overall, miR-150's functions encompass immune regulation, cardioprotection, and modulation of gene expression in various disease contexts.
How ellagic acid activate receptor FXR?
4 answers
Ellagic acid activates the farnesoid X receptor (FXR) through various mechanisms. Studies have shown that ellagic acid promotes cholesterol efflux by upregulating PPARγ and ATP binding cassette transporter-1, crucial for cholesterol efflux in lipid-laden macrophages. Additionally, ellagic acid accelerates the expression and transcription of liver X receptor, highly implicated in PPAR signaling, further enhancing cholesterol efflux in foam cells. Moreover, ellagic acid has been found to protect against inflammation and diarrhea by activating the PPAR signaling pathway, which is associated with FXR activity. These findings suggest that ellagic acid's activation of FXR involves modulating PPAR pathways and promoting cholesterol efflux, highlighting its potential as a therapeutic agent for conditions related to FXR dysregulation.
What is the role of FXR in regulating lipid metabolism?
5 answers
The farnesoid X receptor (FXR) plays a crucial role in regulating lipid metabolism. FXR is involved in the transcriptional regulation of genes related to lipid homeostasis, glucose metabolism, and mitochondrial functions in adipose tissues. Studies have shown that FXR modulates the downstream genes involved in lipid metabolism, such as CYP7A1, impacting lipid accumulation in the liver and preventing liver fibrosis. Additionally, FXR is implicated in the absorption and transportation of lipids through interactions with bile acids, highlighting its significance in lipid metabolic processes. Overall, FXR's regulatory role in lipid metabolism extends to adipose tissue expansion, mitochondrial gene expression, energy expenditure, and systemic glucose tolerance, emphasizing its importance in maintaining whole-body energy homeostasis.
Aminoacyl-tRNA synthetases (ARSs) in breast cancer survival
5 answers
Aminoacyl-tRNA synthetases (ARSs) play crucial roles in protein synthesis and have been implicated in cancer, including breast cancer. Studies have shown that ARSs are often upregulated in tumors, correlating with worse patient survival outcomes. Specifically, methionyl-tRNA synthetase (MARS) has been identified as significantly upregulated in breast cancer tissues, associated with poor prognosis, and linked to cancer progression. The dysregulation of ARSs, including MARS, in breast cancer highlights their potential as prognostic markers and therapeutic targets for improving clinical diagnosis and treatment strategies. Analyzing ARS expression data from large-scale projects like the Cancer Genome Atlas (TCGA) can provide valuable insights into the molecular and prognostic impact of ARSs in breast cancer, potentially identifying specific ARS family members as promising targets for biological cancer therapies.
How does H3K27me repress trasncription?
5 answers
H3K27me3 is involved in transcriptional repression by marking regions associated with gene silencing. This histone modification is catalyzed by the Polycomb Repressive Complex 2 (PRC2), and its global alterations have been linked to various cancer types. In muscle cells, H3K27me3 influences gene expression related to mineral content, impacting muscle function and beef quality. Specifically, H3K27me3 provides significant local transcriptional repression, with regions losing this mark gaining H3K27ac, leading to increased transcription. The dynamic regulation of H3K27 modifications plays a crucial role in fungal genome regulation, especially in the transcriptional control of genes important during host infection. Therefore, H3K27me3 acts as a key epigenetic mark orchestrating transcriptional repression in various biological contexts.
What are the reported functions of CARP2/RFFL with respect to cancer?
5 answers
CARP2/RFFL, a RING-domain E3 ligase, plays crucial roles in cancer progression. It is involved in regulating cell migration, impacting tumor cell survival and sensitivity to apoptosis-inducing agents. CARP2/RFFL is associated with Golgi dynamics through ubiquitination and degradation of Golgi structural proteins like Golgin45, influencing EGF-stimulated cell migration. Additionally, CARP2/RFFL's diverse substrate specificity sets it apart from classical inhibitors of apoptosis, targeting apical caspases for degradation and modulating apoptotic signaling in the extrinsic cell death pathway. These functions highlight the significance of CARP2/RFFL in cancer biology, suggesting its potential as a therapeutic target for intervention in cancer treatment.
What are the specific metabolic pathways that are activated during football training and competition?
5 answers
Football training and competition induce significant metabolic changes in athletes. Metabolomic profiling revealed alterations in pathways such as xanthine, fatty acid (acyl choline), medium chain fatty acid, primary bile acid, and glycolysis, gluconeogenesis, and pyruvate metabolism. Additionally, tyrosine metabolism was notably affected, with changes in homogentisate metabolites, DOPA pathways, and neurotransmitter synthesis. The association between external load and urinary metabolites highlighted steroid hormone biosynthesis, tyrosine, tryptophan metabolism, and riboflavin as key pathways influenced by training. These findings underscore the intricate metabolic adaptations occurring during football activities, emphasizing the importance of understanding and monitoring these pathways for optimizing athlete performance and health.
What controversies surround alpha cell derived GLP-1?
5 answers
Controversies surrounding alpha cell-derived GLP-1 involve its role in paracrine signaling within the pancreatic islets. While the classical incretin model posits that GLP-1 from enteroendocrine L-cells acts on beta-cell GLP-1Rs, recent evidence suggests that alpha-cell-derived GLP-1 may also play a significant role in regulating insulin secretion. This intra-islet crosstalk challenges the traditional understanding of GLP-1's source and function, indicating a potential paracrine signaling pathway from alpha-cells to beta-cells. Additionally, the presence of GLP-1/GLP-1R complexes in diabetic alpha cells suggests a unique endosomal signaling pathway in diabetic conditions, impacting glucagon secretion and contributing to hyperglucagonemia in type 2 diabetes. Further research is needed to fully elucidate the mechanisms and implications of alpha cell-derived GLP-1 in glucose homeostasis.