Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
1.
Islets ; 16(1): 2344622, 2024 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-38652652

RESUMEN

Chronically elevated levels of glucose are deleterious to pancreatic ß cells and contribute to ß cell dysfunction, which is characterized by decreased insulin production and a loss of ß cell identity. The Krüppel-like transcription factor, Glis3 has previously been shown to positively regulate insulin transcription and mutations within the Glis3 locus have been associated with the development of several pathologies including type 2 diabetes mellitus. In this report, we show that Glis3 is significantly downregulated at the transcriptional level in INS1 832/13 cells within hours of being subjected to high glucose concentrations and that diminished expression of Glis3 is at least partly attributable to increased oxidative stress. CRISPR/Cas9-mediated knockdown of Glis3 indicated that the transcription factor was required to maintain normal levels of both insulin and MafA expression and reduced Glis3 expression was concomitant with an upregulation of ß cell disallowed genes. We provide evidence that Glis3 acts similarly to a pioneer factor at the insulin promoter where it permissively remodels the chromatin to allow access to a transcriptional regulatory complex including Pdx1 and MafA. Finally, evidence is presented that Glis3 can positively regulate MafA transcription through its pancreas-specific promoter and that MafA reciprocally regulates Glis3 expression. Collectively, these results suggest that decreased Glis3 expression in ß cells exposed to chronic hyperglycemia may contribute significantly to reduced insulin transcription and a loss of ß cell identity.


Asunto(s)
Regulación hacia Abajo , Glucosa , Células Secretoras de Insulina , Insulina , Proteínas Represoras , Animales , Ratas , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Glucosa/metabolismo , Glucosa/farmacología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Estrés Oxidativo/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
2.
J Biol Chem ; 299(5): 104675, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37028761

RESUMEN

MafA and c-Maf are close members of the Maf transcription factor family and indicators of poor prognosis of multiple myeloma (MM). Our previous study finds that the ubiquitin ligase HERC4 induces c-Maf degradation but stabilizes MafA, and the mechanism is elusive. In the present study, we find that HERC4 interacts with MafA and mediates its K63-linked polyubiquitination at K33. Moreover, HERC4 inhibits MafA phosphorylation and its transcriptional activity triggered by glycogen synthase kinase 3ß (GSK3ß). The K33R MafA variant prevents HERC4 from inhibiting MafA phosphorylation and increases MafA transcriptional activity. Further analyses reveal that MafA can also activate the STAT3 signaling, but it is suppressed by HERC4. Lastly, we demonstrate that lithium chloride, a GSK3ß inhibitor, can upregulate HERC4 and synergizes dexamethasone, a typical anti-MM drug, in inhibiting MM cell proliferation and xenograft growth in nude mice. These findings thus highlight a novel regulation of MafA oncogenic activity in MM and provide the rationale by targeting HERC4/GSK3ß/MafA for the treatment of MM.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta , Factores de Transcripción Maf de Gran Tamaño , Mieloma Múltiple , Poliubiquitina , Ubiquitina-Proteína Ligasas , Ubiquitinación , Animales , Humanos , Ratones , Proliferación Celular , Dexametasona/farmacología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Cloruro de Litio/farmacología , Factores de Transcripción Maf de Gran Tamaño/antagonistas & inhibidores , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Ratones Desnudos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Fosforilación , Poliubiquitina/metabolismo , Factor de Transcripción STAT3/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cell Rep ; 42(4): 112289, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-36952339

RESUMEN

Myofibers are broadly characterized as fatigue-resistant slow-twitch (type I) fibers and rapidly fatiguing fast-twitch (type IIa/IIx/IIb) fibers. However, the molecular regulation of myofiber type is not entirely understood; particularly, information on regulators of fast-twitch muscle is scarce. Here, we demonstrate that the large Maf transcription factor family dictates fast type IIb myofiber specification in mice. Remarkably, the ablation of three large Mafs leads to the drastic loss of type IIb myofibers, resulting in enhanced endurance capacity and the reduction of muscle force. Conversely, the overexpression of each large Maf in the type I soleus muscle induces type IIb myofibers. Mechanistically, a large Maf directly binds to the Maf recognition element on the promoter of myosin heavy chain 4, which encodes the type IIb myosin heavy chain, driving its expression. This work identifies the large Maf transcription factor family as a major regulator for fast type IIb muscle determination.


Asunto(s)
Fibras Musculares de Contracción Rápida , Cadenas Pesadas de Miosina , Ratones , Animales , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Fibras Musculares de Contracción Rápida/metabolismo , Músculo Esquelético/metabolismo , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Proteínas Proto-Oncogénicas c-maf/metabolismo
4.
Endocrinol Metab (Seoul) ; 37(6): 901-917, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36475359

RESUMEN

BACKGRUOUND: Chronic exposure to elevated levels of saturated fatty acids results in pancreatic ß-cell senescence. However, targets and effective agents for preventing stearic acid-induced ß-cell senescence are still lacking. Although melatonin administration can protect ß-cells against lipotoxicity through anti-senescence processes, the precise underlying mechanisms still need to be explored. Therefore, we investigated the anti-senescence effect of melatonin on stearic acid-treated mouse ß-cells and elucidated the possible role of microRNAs in this process. METHODS: ß-Cell senescence was identified by measuring the expression of senescence-related genes and senescence-associated ß-galactosidase staining. Gain- and loss-of-function approaches were used to investigate the involvement of microRNAs in stearic acid-evoked ß-cell senescence and dysfunction. Bioinformatics analyses and luciferase reporter activity assays were applied to predict the direct targets of microRNAs. RESULTS: Long-term exposure to a high concentration of stearic acid-induced senescence and upregulated miR-146a-5p and miR- 8114 expression in both mouse islets and ß-TC6 cell lines. Melatonin effectively suppressed this process and reduced the levels of these two miRNAs. A remarkable reversibility of stearic acid-induced ß-cell senescence and dysfunction was observed after silencing miR-146a-5p and miR-8114. Moreover, V-maf musculoaponeurotic fibrosarcoma oncogene homolog A (Mafa) was verified as a direct target of miR-146a-5p and miR-8114. Melatonin also significantly ameliorated senescence and dysfunction in miR-146a-5pand miR-8114-transfected ß-cells. CONCLUSION: These data demonstrate that melatonin protects against stearic acid-induced ß-cell senescence by inhibiting miR-146a- 5p and miR-8114 and upregulating Mafa expression. This not only provides novel targets for preventing stearic acid-induced ß-cell dysfunction, but also points to melatonin as a promising drug to combat type 2 diabetes progression.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Melatonina , MicroARNs , Ratones , Animales , Melatonina/farmacología , Melatonina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , MicroARNs/farmacología , Senescencia Celular , Ácidos Esteáricos/farmacología , Ácidos Esteáricos/metabolismo , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Factores de Transcripción Maf de Gran Tamaño/farmacología
5.
Int J Mol Sci ; 23(9)2022 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-35562869

RESUMEN

Pancreatic ß-cells are specialized to properly regulate blood glucose. Maintenance of the mature ß-cell phenotype is critical for glucose metabolism, and ß-cell failure results in diabetes mellitus. Recent studies provide strong evidence that the mature phenotype of ß-cells is maintained by several transcription factors. These factors are also required for ß-cell differentiation from endocrine precursors or maturation from immature ß-cells during pancreatic development. Because the reduction or loss of these factors leads to ß-cell failure and diabetes, inducing the upregulation or inhibiting downregulation of these transcription factors would be beneficial for studies in both diabetes and stem cell biology. Here, we discuss one such factor, i.e., the transcription factor MAFA. MAFA is a basic leucine zipper family transcription factor that can activate the expression of insulin in ß-cells with PDX1 and NEUROD1. MAFA is indeed indispensable for the maintenance of not only insulin expression but also function of adult ß-cells. With loss of MAFA in type 2 diabetes, ß-cells cannot maintain their mature phenotype and are dedifferentiated. In this review, we first briefly summarize the functional roles of MAFA in ß-cells and then mainly focus on the molecular mechanism of cell fate conversion regulated by MAFA.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Diferenciación Celular/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo
6.
Biomolecules ; 12(4)2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35454124

RESUMEN

ß-cells are insulin-producing cells in the pancreas that maintain euglycemic conditions. Pancreatic ß-cell maturity and function are regulated by a variety of transcription factors that enable the adequate expression of the cellular machinery involved in nutrient sensing and commensurate insulin secretion. One of the key factors in this regulation is MAF bZIP transcription factor A (MafA). MafA expression is decreased in type 2 diabetes, contributing to ß-cell dysfunction and disease progression. The molecular biology underlying MafA is complex, with numerous transcriptional and post-translational regulatory nodes. Understanding these complexities may uncover potential therapeutic targets to ameliorate ß-cell dysfunction. This article will summarize the role of MafA in normal ß-cell function and disease, with a special focus on known transcriptional and post-translational regulators of MafA expression.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo
7.
Acta Physiol (Oxf) ; 234(2): e13761, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34978761

RESUMEN

AIMS: Reduced expression of exocytotic genes is associated with functional defects in insulin exocytosis contributing to impaired insulin secretion and type 2 diabetes (T2D) development. MAFA and MAFB transcription factors regulate ß-cell physiology, and their gene expression is reduced in T2D ß cells. We investigate if loss of MAFA and MAFB in human ß cells contributes to T2D progression by regulating genes required for insulin exocytosis. METHODS: Three approaches were performed: (1) RNAseq analysis with the focus on exocytosis-related genes in MafA-/- mouse islets, (2) correlational analysis between MAFA, MAFB and exocytosis-related genes in human islets and (3) MAFA and MAFB silencing in human islets and EndoC-ßH1 cells followed by functional in vitro studies. RESULTS: The expression of 30 exocytosis-related genes was significantly downregulated in MafA-/- mouse islets. In human islets, the expression of 29 exocytosis-related genes correlated positively with MAFA and MAFB. Eight exocytosis-related genes were downregulated in MafA-/- mouse islets and positively correlated with MAFA and MAFB in human islets. From this analysis, the expression of RAB3A, STXBP1, UNC13A, VAMP2, NAPA, NSF, STX1A and SYT7 was quantified after acute MAFA or MAFB silencing in EndoC-ßH1 cells and human islets. MAFA and MAFB silencing resulted in impaired insulin secretion and reduced STX1A, SYT7 and STXBP1 (EndoC-ßH1) and STX1A (human islets) mRNA expression. STX1A and STXBP1 protein expression was also impaired in islets from T2D donors which lack MAFA expression. CONCLUSION: Our data indicate that STXBP1 and STX1A are important MAFA/B-regulated exocytosis genes which may contribute to insulin exocytosis defects observed in MAFA-deficient human T2D ß cells.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Animales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Exocitosis , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Factor de Transcripción MafB/genética , Factor de Transcripción MafB/metabolismo , Ratones
8.
Nucleic Acids Res ; 50(11): 6001-6019, 2022 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34897510

RESUMEN

Translational readthrough (TR) occurs when the ribosome decodes a stop codon as a sense codon, resulting in two protein isoforms synthesized from the same mRNA. TR has been identified in several eukaryotic organisms; however, its biological significance and mechanism remain unclear. Here, we quantify TR of several candidate genes in Drosophila melanogaster and characterize the regulation of TR in the large Maf transcription factor Traffic jam (Tj). Using CRISPR/Cas9-generated mutant flies, we show that the TR-generated Tj isoform is expressed in a subset of neural cells of the central nervous system and is excluded from the somatic cells of gonads. Control of TR in Tj is critical for preservation of neuronal integrity and maintenance of reproductive health. The tissue-specific distribution of a release factor splice variant, eRF1H, plays a critical role in modulating differential TR of leaky stop codon contexts. Fine-tuning of gene regulatory functions of transcription factors by TR provides a potential mechanism for cell-specific regulation of gene expression.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción , Animales , Codón de Terminación/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulación de la Expresión Génica , Biosíntesis de Proteínas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
Front Immunol ; 12: 690379, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804002

RESUMEN

Pancreatic ß-cell failure is a critical event in the onset of both main types of diabetes mellitus but underlying mechanisms are not fully understood. ß-cells have low anti-oxidant capacity, making them more susceptible to oxidative stress. In type 1 diabetes (T1D), reactive oxygen species (ROS) are associated with pro-inflammatory conditions at the onset of the disease. Here, we investigated the effects of hydrogen peroxide-induced oxidative stress on human ß-cells. We show that primary human ß-cell function is decreased. This reduced function is associated with an ER stress response and the shuttling of FOXO1 to the nucleus. Furthermore, oxidative stress leads to loss of ß-cell maturity genes MAFA and PDX1, and to a concomitant increase in progenitor marker expression of SOX9 and HES1. Overall, we propose that oxidative stress-induced ß-cell failure may result from partial dedifferentiation. Targeting antioxidant mechanisms may preserve functional ß-cell mass in early stages of development of T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Estrés Oxidativo/fisiología , Antioxidantes/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/fisiopatología , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción SOX9/metabolismo , Transactivadores/metabolismo , Factor de Transcripción HES-1/metabolismo
10.
Cell Rep ; 37(2): 109813, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34644565

RESUMEN

A heterozygous missense mutation of the islet ß cell-enriched MAFA transcription factor (p.Ser64Phe [S64F]) is found in patients with adult-onset ß cell dysfunction (diabetes or insulinomatosis), with men more prone to diabetes than women. This mutation engenders increased stability to the unstable MAFA protein. Here, we develop a S64F MafA mouse model to determine how ß cell function is affected and find sex-dependent phenotypes. Heterozygous mutant males (MafAS64F/+) display impaired glucose tolerance, while females are slightly hypoglycemic with improved blood glucose clearance. Only MafAS64F/+ males show transiently higher MafA protein levels preceding glucose intolerance and sex-dependent changes to genes involved in Ca2+ signaling, DNA damage, aging, and senescence. MAFAS64F production in male human ß cells also accelerate cellular senescence and increase senescence-associated secretory proteins compared to cells expressing MAFAWT. These results implicate a conserved mechanism of accelerated islet aging and senescence in promoting diabetes in MAFAS64F carriers in a sex-biased manner.


Asunto(s)
Senescencia Celular , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Animales , Animales Modificados Genéticamente , Glucemia/metabolismo , Señalización del Calcio , Línea Celular , Daño del ADN , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Humanos , Insulina/sangre , Células Secretoras de Insulina/patología , Factores de Transcripción Maf de Gran Tamaño/genética , Masculino , Ratones Endogámicos C57BL , Mutación Missense , Fenotipo , Caracteres Sexuales , Factores Sexuales
11.
JCI Insight ; 6(18)2021 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-34428183

RESUMEN

Islet-enriched transcription factors (TFs) exert broad control over cellular processes in pancreatic α and ß cells, and changes in their expression are associated with developmental state and diabetes. However, the implications of heterogeneity in TF expression across islet cell populations are not well understood. To define this TF heterogeneity and its consequences for cellular function, we profiled more than 40,000 cells from normal human islets by single-cell RNA-Seq and stratified α and ß cells based on combinatorial TF expression. Subpopulations of islet cells coexpressing ARX/MAFB (α cells) and MAFA/MAFB (ß cells) exhibited greater expression of key genes related to glucose sensing and hormone secretion relative to subpopulations expressing only one or neither TF. Moreover, all subpopulations were identified in native pancreatic tissue from multiple donors. By Patch-Seq, MAFA/MAFB-coexpressing ß cells showed enhanced electrophysiological activity. Thus, these results indicate that combinatorial TF expression in islet α and ß cells predicts highly functional, mature subpopulations.


Asunto(s)
Células Secretoras de Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Adulto , Fenómenos Electrofisiológicos , Expresión Génica , Células Secretoras de Glucagón/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/fisiología , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Factor de Transcripción MafB/genética , Factor de Transcripción MafB/metabolismo , Persona de Mediana Edad , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transcriptoma , Adulto Joven
12.
J Mol Endocrinol ; 67(3): 71-82, 2021 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-34223824

RESUMEN

Glucose transporter type 2 (GLUT2), encoded by the SLC2A2 gene, is an essential component of glucose-stimulated insulin secretion in pancreatic islet ß-cells. Like that of the gene encoding insulin, expression of the SLC2A2 gene expression is closely linked to ß-cell functionality in rodents, but the mechanism by which ß-cell-specific expression of SLC2A2 is controlled remains unclear. In this report, to identify putative enhancer elements of the mouse Slc2a2 gene, we examined evolutional conservation of the nucleotide sequence of its genomic locus, together with ChIP-seq data of histone modifications and various transcription factors published in previous studies. Using luciferase reporter assays, we found that an evolutionarily conserved region (ECR) located approximately 40 kbp downstream of the transcription start site of Slc2a2 functions as an active enhancer in the MIN6 ß-cell line. We also found that three ß-cell-enriched transcription factors, MafA, NeuroD1, and HNF1ß, synergistically activate transcription through this 3' downstream distal enhancer (ECR3') and the proximal promoter region of the gene. Our data also indicate that the simultaneous binding of HNF1ß to its target sites within the promoter and ECR3' of Slc2a2 is indispensable for transcriptional activation, and that binding of MafA and NeuroD1 to their respective target sites within the ECR3' enhances transcription. Co-immunoprecipitation experiments suggested that MafA, NeuroD1, and HNF1ß interact with each other. Overall, these results suggest that promoter-enhancer communication through MafA, NeuroD1, and HNF1ß is critical for Slc2a2 gene expression. These findings provide clues to help elucidate the mechanism of regulation of Slc2a2 gene expression in ß-cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 2/genética , Factor Nuclear 1-beta del Hepatocito/metabolismo , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Animales , Secuencia Conservada , Elementos de Facilitación Genéticos , Transportador de Glucosa de Tipo 2/metabolismo , Ratones , Regiones Promotoras Genéticas , Unión Proteica , Elementos de Respuesta , Activación Transcripcional
13.
Biochem Biophys Res Commun ; 556: 134-141, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33839409

RESUMEN

Oxidative stress is a deteriorating factor for pancreatic ß-cells under chronic hyperglycemia in diabetes. However, the molecular mechanism underlying the increase in oxidative stress in ß-cells under diabetic conditions remains unclear. We demonstrated previously that the selective alleviation of glucotoxicity ameliorated the downregulation of several ß-cell factors, including Cox6a2. Cox6a2 encodes a subunit of the respiratory chain complex IV in mitochondria. In this study, we analyzed the role of Cox6a2 in pancreatic ß-cell function and its pathophysiological significance in diabetes mellitus. Cox6a2-knockdown experiments in MIN6-CB4 cells indicated an increased production of reactive oxygen species as detected by CellROX Deep Red reagent using flow cytometry. In systemic Cox6a2-knockout mice, impaired glucose tolerance was observed under a high-fat high-sucrose diet. However, insulin resistance was reduced when compared with control littermates. This indicates a relative insufficiency of ß-cell function. To examine the transcriptional regulation of Cox6a2, ATAC-seq with islet DNA was performed and an open-chromatin area within the Cox6a2 enhancer region was detected. Reporter gene analysis using this area revealed that MafA directly regulates Cox6a2 expression. These findings suggest that the decreased expression of Cox6a2 increases the levels of reactive oxygen species and that Mafa is associated with decreased Cox6a2 expression under glucotoxic conditions.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Proteínas Musculares/deficiencia , Especies Reactivas de Oxígeno/metabolismo , Animales , Línea Celular , Diabetes Mellitus Experimental/metabolismo , Complejo IV de Transporte de Electrones/biosíntesis , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Regulación de la Expresión Génica , Glucosa/metabolismo , Intolerancia a la Glucosa/genética , Células HEK293 , Humanos , Insulina/metabolismo , Resistencia a la Insulina/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Estrés Oxidativo , Transcripción Genética
14.
Int J Mol Sci ; 22(5)2021 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-33801253

RESUMEN

P43 is a truncated form of thyroid hormone receptor α localized in mitochondria, which stimulates mitochondrial respiratory chain activity. Previously, we showed that deletion of p43 led to reduction of pancreatic islet density and a loss of glucose-stimulated insulin secretion in adult mice. The present study was designed to determine whether p43 was involved in the processes of ß cell development and maturation. We used neonatal, juvenile, and adult p43-/- mice, and we analyzed the development of ß cells in the pancreas. Here, we show that p43 deletion affected only slightly ß cell proliferation during the postnatal period. However, we found a dramatic fall in p43-/- mice of MafA expression (V-Maf Avian Musculoaponeurotic Fibrosarcoma Oncogene Homolog A), a key transcription factor of beta-cell maturation. Analysis of the expression of antioxidant enzymes in pancreatic islet and 4-hydroxynonenal (4-HNE) (a specific marker of lipid peroxidation) staining revealed that oxidative stress occurred in mice lacking p43. Lastly, administration of antioxidants cocktail to p43-/- pregnant mice restored a normal islet density but failed to ensure an insulin secretion in response to glucose. Our findings demonstrated that p43 drives the maturation of ß cells via its induction of transcription factor MafA during the critical postnatal window.


Asunto(s)
Diferenciación Celular , Regulación de la Expresión Génica , Secreción de Insulina , Células Secretoras de Insulina/citología , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Receptores alfa de Hormona Tiroidea/fisiología , Animales , Femenino , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Masculino , Ratones , Ratones Noqueados , Estrés Oxidativo
15.
Exp Anim ; 70(3): 264-271, 2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-33762508

RESUMEN

The large MAF transcription factor group is a group of transcription factors with an acidic region, a basic region, and a leucine zipper region. Four types of MAF, MAFA, MAFB, c-MAF, and NRL, have been identified in humans and mice. In order to elucidate the functions of the large MAF transcription factor group in vivo, our research group created genetically modified MAFA-, MAFB-, and c-MAF-deficient mice and analyzed their phenotypes. MAFA is expressed in pancreatic ß cells and is essential for insulin transcription and secretion. MAFB is essential for the development of pancreatic endocrine cells, formation of inner ears, podocyte function in the kidneys, and functional differentiation of macrophages. c-MAF is essential for lens formation and osteoblast differentiation. Furthermore, a single-base mutation in genes encoding the large MAF transcription factor group causes congenital renal disease, eye disease, bone disease, diabetes, and tumors in humans. This review describes the functions of large MAF transcription factors in vivo and their relationships with human diseases.


Asunto(s)
Enfermedades Óseas/genética , Diabetes Mellitus/genética , Oftalmopatías/genética , Enfermedades Renales/genética , Factores de Transcripción Maf de Gran Tamaño/genética , Mutación , Neoplasias/genética , Animales , Oftalmopatías/congénito , Humanos , Enfermedades Renales/congénito , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Ratones
16.
Diabetes ; 70(5): 1117-1122, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33685924

RESUMEN

Single-cell RNA-sequencing (scRNA-Seq) technologies have greatly enhanced our understanding of islet cell transcriptomes and have revealed the existence of ß-cell heterogeneity. However, comparison of scRNA-Seq data sets from different groups have highlighted inconsistencies in gene expression patterns, primarily due to variable detection of lower abundance transcripts. Furthermore, such analyses are unable to uncover the spatial organization of heterogeneous gene expression. In this study, we used fluctuation localization imaging-based fluorescence in situ hybridization (fliFISH) to quantify transcripts in single cells in mouse pancreatic islet sections. We compared the expression patterns of Insulin 2 (Ins2) with Mafa and Ucn3, two genes expressed in ß-cells as they mature, as well as Rgs4, a factor with variably reported expression in the islet. This approach accurately quantified transcripts across a wide range of expression levels, from single copies to >100 copies/cell in one islet. Importantly, fliFISH allowed evaluation of transcript heterogeneity in the spatial context of an intact islet. These studies confirm the existence of a high degree of heterogeneous gene expression levels within the islet and highlight relative and radial expression patterns that likely reflect distinct ß-cell maturation states along the radial axis of the islet.


Asunto(s)
Células Secretoras de Insulina/metabolismo , RNA-Seq/métodos , Análisis de Secuencia de ARN/métodos , Animales , Hibridación Fluorescente in Situ , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Ratones , Proteínas RGS/genética , Proteínas RGS/metabolismo , Análisis de la Célula Individual , Urocortinas/genética , Urocortinas/metabolismo
17.
Metabolism ; 118: 154734, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33631146

RESUMEN

The amplification of glucose-stimulated insulin secretion (GSIS) through incretin signaling is critical for maintaining physiological glucose levels. Incretins, like glucagon-like peptide 1 (GLP1), are a target of type 2 diabetes drugs aiming to enhance insulin secretion. Here we show that the protein phosphatase 1 inhibitor protein 1A (PPP1R1A), is expressed in ß-cells and that its expression is reduced in dysfunctional ß-cells lacking MafA and upon acute MafA knock down. MafA is a central regulator of GSIS and ß-cell function. We observed a strong correlation of MAFA and PPP1R1A mRNA levels in human islets, moreover, PPP1R1A mRNA levels were reduced in type 2 diabetic islets and positively correlated with GLP1-mediated GSIS amplification. PPP1R1A silencing in INS1 (832/13) ß-cells impaired GSIS amplification, PKA-target protein phosphorylation, mitochondrial coupling efficiency and also the expression of critical ß-cell marker genes like MafA, Pdx1, NeuroD1 and Pax6. Our results demonstrate that the ß-cell transcription factor MafA is required for PPP1R1A expression and that reduced ß-cell PPP1R1A levels impaired ß-cell function and contributed to ß-cell dedifferentiation during type 2 diabetes. Loss of PPP1R1A in type 2 diabetic ß-cells may explains the unresponsiveness of type 2 diabetic patients to GLP1R-based treatments.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Proteína Fosfatasa 1/genética , Animales , Desdiferenciación Celular , Línea Celular , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Humanos , Células Secretoras de Insulina/patología , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Fosforilación , ARN Mensajero/genética
18.
Nat Commun ; 12(1): 674, 2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33514698

RESUMEN

Transcriptionally mature and immature ß-cells co-exist within the adult islet. How such diversity contributes to insulin release remains poorly understood. Here we show that subtle differences in ß-cell maturity, defined using PDX1 and MAFA expression, contribute to islet operation. Functional mapping of rodent and human islets containing proportionally more PDX1HIGH and MAFAHIGH ß-cells reveals defects in metabolism, ionic fluxes and insulin secretion. At the transcriptomic level, the presence of increased numbers of PDX1HIGH and MAFAHIGH ß-cells leads to dysregulation of gene pathways involved in metabolic processes. Using a chemogenetic disruption strategy, differences in PDX1 and MAFA expression are shown to depend on islet Ca2+ signaling patterns. During metabolic stress, islet function can be restored by redressing the balance between PDX1 and MAFA levels across the ß-cell population. Thus, preserving heterogeneity in PDX1 and MAFA expression, and more widely in ß-cell maturity, might be important for the maintenance of islet function.


Asunto(s)
Secreción de Insulina/fisiología , Células Secretoras de Insulina/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Femenino , Técnicas de Sustitución del Gen , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Masculino , Ratones , Ratones Transgénicos , Modelos Animales , Cultivo Primario de Células , Transactivadores/genética , Transactivadores/metabolismo
19.
Pancreas ; 50(10): 1450-1453, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35041347

RESUMEN

ABSTRACT: Sporadic adult insulinomatosis is an extremely rare clinical condition. Adult proinsulinomatosis has not yet been described. We report the case of a 48-year-old female patient with recurrent hypoglycemia caused by benign proinsulin-secreting pancreatic neuroendocrine neoplasias (pNENs) with no history of multiple endocrine neoplasia type 1. Initial workup revealed elevated serum proinsulin levels and a positive fasting test. Magnetic resonance imaging and endosonography visualized 2 pNENs in the pancreatic body and tail that were treated by robotic-assisted enucleation. After initial biochemical cure, the patient's hypoglycemia recurred 3 months after surgery. Imaging showed a new lesion in the pancreatic body, so that now a spleen-preserving subtotal distal pancreatectomy was performed. The pathological examination revealed 17 neuroendocrine microadenomas and 1 well-differentiated pNEN (Ki-67% 1%-2%) of 22-mm size as well as more than 200 (pro)insulin-producing ß-cell precursor lesions, confirming the diagnosis of adult proinsulinomatosis. Mutation analysis of the germline DNA identified the in-frame deletion mutation (p.His207del) in the MAFA gene on chromosome 8. The patient was biochemically cured 16 months after the last surgical resection. Similarly to adult insulinomatosis, the presence of proinsulin-secreting tumors causes recurrent hypoglycemia and might be associated with germline mutations in the MAFA gene.


Asunto(s)
Hipoglucemia/etiología , Insulinoma/complicaciones , Factores de Transcripción Maf de Gran Tamaño/genética , Glucemia/análisis , Glucemia/biosíntesis , Femenino , Alemania , Humanos , Hipoglucemia/genética , Insulinoma/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Persona de Mediana Edad , Mutación/genética , Proinsulina/sangre
20.
Virulence ; 11(1): 1701-1715, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33315509

RESUMEN

MafB proteins are toxins secreted by Neisseria spp. which are involved in interbacterial competition. Their secretion mechanism has so far not been elucidated. Each strain can produce several MafB variants. On the chromosome, the mafB genes are localized on genomic islands also containing mafA genes. MafA proteins have a role in virulence with reported activities in adhesion and transcytosis of pathogenic Neisseria, a priori unrelated to MafB activities. In this study, we investigated the possible involvement of MafA in the transport of MafB across the outer membrane of Neisseria meningitidis. In wild-type strains, proteolytic fragments of MafB proteins were detected in the extracellular medium. In the absence of MafA, secretion was abrogated, and, in the case of MafBI, full-length and truncated polypeptides were detected inside the cells and inside outer-membrane vesicles. MafBI secretion required its cognate MafA, whereas MafBIII could use any MafA. Heterologous expression in Escherichia coli showed that MafBIII is transported to a cell-surface-exposed, i.e. protease-accessible, location in a MafA-dependent way. MafA itself was found to be localized to the outer membrane, forming large oligomeric complexes. As homologs were found in diverse bacteria, the Maf system represents a new protein secretion system in Gram-negative bacteria.


Asunto(s)
Membrana Externa Bacteriana/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Factor de Transcripción MafB/genética , Neisseria meningitidis/genética , Neisseria meningitidis/metabolismo , Vías Secretoras , Membrana Externa Bacteriana/química , Transporte Biológico , Escherichia coli/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Factor de Transcripción MafB/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...