Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Development ; 149(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35708349

ABSTRACT

Pancreatic and duodenal homeobox 1 (PDX1) is crucial for pancreas organogenesis, yet the dynamic changes in PDX1 binding in human or mouse developing pancreas have not been examined. To address this knowledge gap, we performed PDX1 ChIP-seq and single-cell RNA-seq using fetal human pancreata. We integrated our datasets with published datasets and revealed the dynamics of PDX1 binding and potential cell lineage-specific PDX1-bound genes in the pancreas from fetal to adult stages. We identified a core set of developmentally conserved PDX1-bound genes that reveal the broad multifaceted role of PDX1 in pancreas development. Despite the well-known dramatic changes in PDX1 function and expression, we found that PDX1-bound genes are largely conserved from embryonic to adult stages. This points towards a dual role of PDX1 in regulating the expression of its targets at different ages, dependent on other functionally congruent or directly interacting partners. We also showed that PDX1 binding is largely conserved in mouse pancreas. Together, our study reveals PDX1 targets in the developing pancreas in vivo and provides an essential resource for future studies on pancreas development.


Subject(s)
Genes, Homeobox , Homeodomain Proteins , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Pancreas , Trans-Activators/genetics , Trans-Activators/metabolism , Transcriptome/genetics
2.
Aesthet Surg J ; 40(1): 108-117, 2020 01 01.
Article in English | MEDLINE | ID: mdl-30873530

ABSTRACT

BACKGROUND: The efficacy of adipose-derived stem cells (ASCs) to improve wound healing has been extensively investigated. Unfortunately, no consistent reports have described significant improvements in healing time or outcomes in large-scale clinical trials utilizing human ASCs. Primarily, these studies examined undifferentiated ASCs as opposed to specific cells differentiated from ASCs. OBJECTIVES: The authors sought to examine the properties of fibroblasts differentiated from human ASCs (dFib cells) compared with those of primary dermal fibroblasts. METHODS: ASCs were isolated from healthy female patients, differentiated into dFib cells, and compared with intra-patient primary dermal fibroblasts for morphology, extracellular matrix (ECM) marker expression, and cell migration employing qPCR, western blot, and scratch test assays. RESULTS: De novo differentiated fibroblasts produce higher levels of the healthy ECM markers Elastin, Fibronectin, and Collagen 1 compared with primary fibroblasts. In contrast, dFib cells have reduced expression of the scar tissue markers αSMA, Collagen 3, and MMP-1. Further, dFib cells close scratch defects more quickly than primary dermal fibroblasts (32 ± 12.85 hours vs 64 ± 13.85 hours, P < 0.01) in a scratch test assay. CONCLUSIONS: These data suggest that fibroblasts newly differentiated from human ASCs migrate well and produce a robust ECM, the combination of which may contribute to improved wound healing, and thus should be further investigated.


Subject(s)
Adipose Tissue , Stem Cells , Adipocytes , Extracellular Matrix , Female , Fibroblasts , Humans
3.
Science ; 348(6242): aaa6071, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-26113728

ABSTRACT

Cardiac progenitor cells are multipotent and give rise to cardiac endothelium, smooth muscle, and cardiomyocytes. Here, we define and characterize the cardiomyoblast intermediate that is committed to the cardiomyocyte fate, and we characterize the niche signals that regulate commitment. Cardiomyoblasts express Hopx, which functions to coordinate local Bmp signals to inhibit the Wnt pathway, thus promoting cardiomyogenesis. Hopx integrates Bmp and Wnt signaling by physically interacting with activated Smads and repressing Wnt genes. The identification of the committed cardiomyoblast that retains proliferative potential will inform cardiac regenerative therapeutics. In addition, Bmp signals characterize adult stem cell niches in other tissues where Hopx-mediated inhibition of Wnt is likely to contribute to stem cell quiescence and to explain the role of Hopx as a tumor suppressor.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Gene Expression Regulation, Developmental , Heart/embryology , Homeodomain Proteins/metabolism , Myoblasts, Cardiac/metabolism , Organogenesis/genetics , Tumor Suppressor Proteins/metabolism , Wnt Signaling Pathway/genetics , Amino Acid Sequence , Animals , Bone Morphogenetic Proteins/genetics , Cell Lineage/genetics , Gene Expression , Homeodomain Proteins/genetics , Mice , Mice, Mutant Strains , Molecular Sequence Data , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Myoblasts, Cardiac/cytology , Stem Cell Niche/genetics , Stem Cell Niche/physiology , Tumor Suppressor Proteins/genetics
4.
Diabetes ; 64(10): 3475-84, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26085571

ABSTRACT

Mitophagy is a critical regulator of mitochondrial quality control and is necessary for elimination of dysfunctional mitochondria to maintain cellular respiration. Here, we report that the homeodomain transcription factor Pdx1, a gene associated with both type 2 diabetes and monogenic diabetes of the young, regulates mitophagy in pancreatic ß-cells. Loss of Pdx1 leads to abnormal mitochondrial morphology and function as well as impaired mitochondrial turnover. High-throughput expression microarray and chromatin occupancy analyses reveal that Pdx1 regulates the expression of Clec16a, a type 1 diabetes gene and itself a key mediator of mitophagy through regulation of the E3 ubiquitin ligase Nrdp1. Indeed, expression of Clec16a and Nrdp1 are both reduced in Pdx1 haploinsufficient islets, and reduction of Pdx1 impairs fusion of autophagosomes containing mitochondria to lysosomes during mitophagy. Importantly, restoration of Clec16a expression after Pdx1 loss of function restores mitochondrial trafficking during mitophagy and improves mitochondrial respiration and glucose-stimulated insulin release. Thus, Pdx1 orchestrates nuclear control of mitochondrial function in part by controlling mitophagy through Clec16a. The novel Pdx1-Clec16a-Nrdp1 pathway we describe provides a genetic basis for the pathogenesis of mitochondrial dysfunction in multiple forms of diabetes that could be targeted for future therapies to improve ß-cell function.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation/physiology , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/physiology , Lectins, C-Type/metabolism , Mitophagy/physiology , Monosaccharide Transport Proteins/metabolism , Trans-Activators/metabolism , Animals , Carrier Proteins/genetics , DNA/genetics , DNA/metabolism , Homeodomain Proteins/genetics , Humans , Lectins, C-Type/genetics , Mice , Mitochondria/physiology , Monosaccharide Transport Proteins/genetics , Protein Array Analysis , RNA/genetics , RNA/metabolism , Trans-Activators/genetics , Ubiquitin-Protein Ligases
5.
Cell Stem Cell ; 16(4): 386-99, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25842977

ABSTRACT

Embryonic development relies on the capacity of progenitor cells to appropriately respond to inductive cues, a cellular property known as developmental competence. Here, we report that epigenetic priming of enhancers signifies developmental competence during endodermal lineage diversification. Chromatin mapping during pancreatic and hepatic differentiation of human embryonic stem cells revealed the en masse acquisition of a poised chromatin state at enhancers specific to endoderm-derived cell lineages in gut tube intermediates. Experimentally, the acquisition of this poised enhancer state predicts the ability of endodermal intermediates to respond to inductive signals. Furthermore, these enhancers are first recognized by the pioneer transcription factors FOXA1 and FOXA2 when competence is acquired, while subsequent recruitment of lineage-inductive transcription factors, such as PDX1, leads to enhancer and target gene activation. Together, our results identify the acquisition of a poised chromatin state at enhancers as a mechanism by which progenitor cells acquire developmental competence.


Subject(s)
Embryonic Stem Cells/physiology , Endoderm/physiology , Enhancer Elements, Genetic/genetics , Gastrula/physiology , Pancreas/physiology , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Line , Cell Lineage/genetics , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , High-Throughput Nucleotide Sequencing , Histones/genetics , Histones/metabolism , Homeodomain Proteins/metabolism , Humans , Organ Specificity/genetics , RNA, Small Interfering/genetics , Trans-Activators/metabolism
6.
Diabetes ; 64(8): 2905-14, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25918232

ABSTRACT

The homeodomain transcription factor Pdx1 controls pancreas organogenesis, specification of endocrine pancreas progenitors, and the postnatal growth and function of pancreatic ß-cells. Pdx1 expression in human-derived stem cells is used as a marker for induced pancreatic precursor cells. Unfortunately, the differentiation efficiency of human pancreatic progenitors into functional ß-cells is poor. In order to gain insight into the genes that Pdx1 regulates during differentiation, we performed Pdx1 chromatin immunoprecipitation followed by high-throughput sequencing of embryonic day (e) 13.5 and 15.5 mouse pancreata. From this, we identified the transcription factor Teashirt zinc finger 1 (Tshz1) as a direct Pdx1 target. Tshz1 is expressed in developing and adult insulin- and glucagon-positive cells. Endocrine cells are properly specified in Tshz1-null embryos, but critical regulators of ß-cell (Pdx1 and Nkx6.1) and α-cell (MafB and Arx) formation and function are downregulated. Adult Tshz1(+/-) mice display glucose intolerance due to defects in glucose-stimulated insulin secretion associated with reduced Pdx1 and Clec16a expression in Tshz1(+/-) islets. Lastly, we demonstrate that TSHZ1 levels are reduced in human islets of donors with type 2 diabetes. Thus, we position Tshz1 in the transcriptional network of maturing ß-cells and suggest that its dysregulation could contribute to the islet phenotype of human type 2 diabetes.


Subject(s)
Cell Differentiation/genetics , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/metabolism , Organogenesis/genetics , Pancreas/metabolism , Repressor Proteins/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Gene Regulatory Networks , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/cytology , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Pancreas/cytology , Repressor Proteins/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
7.
Proc Natl Acad Sci U S A ; 111(22): E2319-28, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24843127

ABSTRACT

Insulin resistance, hyperinsulinemia, and hyperproinsulinemia occur early in the pathogenesis of type 2 diabetes (T2D). Elevated levels of proinsulin and proinsulin intermediates are markers of ß-cell dysfunction and are strongly associated with development of T2D in humans. However, the mechanism(s) underlying ß-cell dysfunction leading to hyperproinsulinemia is poorly understood. Here, we show that disruption of insulin receptor (IR) expression in ß cells has a direct impact on the expression of the convertase enzyme carboxypeptidase E (CPE) by inhibition of the eukaryotic translation initiation factor 4 gamma 1 translation initiation complex scaffolding protein that is mediated by the key transcription factors pancreatic and duodenal homeobox 1 and sterol regulatory element-binding protein 1, together leading to poor proinsulin processing. Reexpression of IR or restoring CPE expression each independently reverses the phenotype. Our results reveal the identity of key players that establish a previously unknown link between insulin signaling, translation initiation, and proinsulin processing, and provide previously unidentified mechanistic insight into the development of hyperproinsulinemia in insulin-resistant states.


Subject(s)
Carboxypeptidase H/metabolism , Diabetes Mellitus, Type 2/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Insulin-Secreting Cells/physiology , Insulin/metabolism , Animals , Carboxypeptidase H/genetics , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Endoplasmic Reticulum Stress/physiology , Eukaryotic Initiation Factor-4G/genetics , Genome-Wide Association Study , Homeodomain Proteins/metabolism , Humans , Insulin-Secreting Cells/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Biosynthesis/physiology , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction/physiology , Sterol Regulatory Element Binding Protein 1/metabolism , Trans-Activators/metabolism
8.
Mol Endocrinol ; 25(2): 339-47, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21193557

ABSTRACT

The expression pattern of genes important for pancreatic islet cell function requires the actions of cell-enriched transcription factors. Musculoaponeurotic fibrosarcoma homolog A (MafA) is a ß-cell-specific transcriptional activator critical to adult islet ß-cell function, with MafA mutant mice manifesting symptoms associated with human type 2 diabetes. Here, we describe that MafA expression is controlled by hepatocyte nuclear factor 1-α (Hnf1α), the transcription factor gene mutated in the most common monoallelic form of maturity onset diabetes of the young. There are six conserved sequence domains in the 5'-flanking MafA promoter, of which one, region 3 (R3) [base pair (bp) -8118/-7750] is principally involved in controlling the unique developmental and adult islet ß-cell-specific expression pattern. Chromatin immunoprecipitation analysis demonstrated that Hnf1α bound specifically within R3. Furthermore, in vitro DNA-binding experiments localized an Hnf1α regulatory element between bp -7822 and -7793, an area previously associated with stimulation by the islet developmental regulator, Islet1. However, site-directed mutational studies showed that Hnf1α was essential to R3-driven reporter activation through bp -7816/-7811. Significantly, MafA levels were dramatically reduced in the insulin(+) cell population remaining in embryonic and adult Hnf1α(-/-) pancreata. Our results demonstrate that Hnf1α regulates MafA in ß-cells and suggests that compromised MafA expression contributes to ß-cell dysfunction in maturity onset diabetes of the young.


Subject(s)
Hepatocyte Nuclear Factor 1-alpha/metabolism , Insulin-Secreting Cells/metabolism , Maf Transcription Factors, Large/genetics , Animals , Animals, Genetically Modified , Blotting, Western , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Electrophoretic Mobility Shift Assay , Gene Expression , Hepatocyte Nuclear Factor 1-alpha/genetics , Humans , Maf Transcription Factors, Large/metabolism , Mice , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid
9.
J Biol Chem ; 285(51): 40050-9, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-20943662

ABSTRACT

The calcium-regulated phosphatase calcineurin intersects with both calcium and cAMP-mediated signaling pathways in the pancreatic ß-cell. Pharmacologic calcineurin inhibition, necessary to prevent rejection in the setting of organ transplantation, is associated with post-transplant ß-cell failure. We sought to determine the effect of calcineurin inhibition on ß-cell replication and survival in rodents and in isolated human islets. Further, we assessed whether the GLP-1 receptor agonist and cAMP stimulus, exendin-4 (Ex-4), could rescue ß-cell replication and survival following calcineurin inhibition. Following treatment with the calcineurin inhibitor tacrolimus, human ß-cell apoptosis was significantly increased. Although we detected no human ß-cell replication, tacrolimus significantly decreased rodent ß-cell replication. Ex-4 nearly normalized both human ß-cell survival and rodent ß-cell replication when co-administered with tacrolimus. We found that tacrolimus decreased Akt phosphorylation, suggesting that calcineurin could regulate replication and survival via the PI3K/Akt pathway. We identify insulin receptor substrate-2 (Irs2), a known cAMP-responsive element-binding protein target and upstream regulator of the PI3K/Akt pathway, as a novel calcineurin target in ß-cells. Irs2 mRNA and protein are decreased by calcineurin inhibition in both rodent and human islets. The effect of calcineurin on Irs2 expression is mediated at least in part through the nuclear factor of activated T-cells (NFAT), as NFAT occupied the Irs2 promoter in a calcineurin-sensitive manner. Ex-4 restored Irs2 expression in tacrolimus-treated rodent and human islets nearly to baseline. These findings reveal calcineurin as a regulator of human ß-cell survival in part through regulation of Irs2, with implications for the pathogenesis and treatment of diabetes following organ transplantation.


Subject(s)
Calcineurin/pharmacology , Cell Proliferation/drug effects , Insulin-Secreting Cells/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Calcineurin/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cyclic AMP/metabolism , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Exenatide , Gene Expression Regulation/drug effects , Glucagon-Like Peptide-1 Receptor , Humans , Hypoglycemic Agents/pharmacology , Immunosuppressive Agents/pharmacology , Insulin Receptor Substrate Proteins/metabolism , Insulin-Secreting Cells/pathology , Mice , NFATC Transcription Factors/metabolism , Organ Transplantation/adverse effects , Peptides/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Tacrolimus/pharmacology , Venoms/pharmacology
10.
Mol Cell Biol ; 30(17): 4234-44, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20584984

ABSTRACT

MafA is a key transcriptional activator of islet beta cells, and its exclusive expression within beta cells of the developing and adult pancreas is distinct among pancreatic regulators. Region 3 (base pairs -8118 to -7750 relative to the transcription start site), one of six conserved 5' cis domains of the MafA promoter, is capable of directing beta-cell-line-selective expression. Transgenic reporters of region 3 alone (R3), sequences spanning regions 1 to 6 (R1-6; base pairs -10428 to +230), and R1-6 lacking R3 (R1-6(DeltaR3)) were generated. Only the R1-6 transgene was active in MafA(+) insulin(+) cells during development and in adult cells. R1-6 also mediated glucose-induced MafA expression. Conversely, pancreatic expression was not observed with the R3 or R1-6(DeltaR3) line, although much of the nonpancreatic expression pattern was shared between the R1-6 and R1-6(DeltaR3) lines. Further support for the importance of R3 was also shown, as the islet regulators Nkx6.1 and Pax6, but not NeuroD1, activated MafA in gel shift, chromatin immunoprecipitation (ChIP), and transfection assays and in vivo mouse knockout models. Lastly, ChIP demonstrated that Pax6 and Pdx-1 also bound to R1 and R6, potentially functioning in pancreatic and nonpancreatic expression. These data highlight the nature of the cis- and trans-acting factors controlling the beta-cell-specific expression of MafA.


Subject(s)
5' Flanking Region , Insulin-Secreting Cells/metabolism , Maf Transcription Factors, Large/genetics , Transcription, Genetic , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Eye Proteins/metabolism , Glucose/metabolism , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/metabolism , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Protein Binding , Regulatory Sequences, Nucleic Acid , Repressor Proteins/metabolism
11.
Proc Natl Acad Sci U S A ; 104(10): 3853-8, 2007 Mar 06.
Article in English | MEDLINE | ID: mdl-17360442

ABSTRACT

Pancreatic endocrine cell differentiation depends on transcription factors that also contribute in adult insulin and glucagon gene expression. Islet cell development was examined in mice lacking MafB, a transcription factor expressed in immature alpha (glucagon(+)) and beta (insulin(+)) cells and capable of activating insulin and glucagon expression in vitro. We observed that MafB(-/-) embryos had reduced numbers of insulin(+) and glucagon(+) cells throughout development, whereas the total number of endocrine cells was unchanged. Moreover, production of insulin(+) cells was delayed until embryonic day (E) 13.5 in mutant mice and coincided with the onset of MafA expression, a MafB-related activator of insulin transcription. MafA expression was only detected in the insulin(+) cell population in MafB mutants, whereas many important regulatory proteins continued to be expressed in insulin(-) beta cells. However, Pdx1, Nkx6.1, and GLUT2 were selectively lost in these insulin-deficient cells between E15.5 and E18.5. MafB appears to directly regulate transcription of these genes, because binding was observed within endogenous control region sequences. These results demonstrate that MafB plays a previously uncharacterized role by regulating transcription of key factors during development that are required for the production of mature alpha and beta cells.


Subject(s)
Insulin-Secreting Cells/metabolism , MafB Transcription Factor/genetics , MafB Transcription Factor/physiology , Animals , Cell Differentiation , Glucagon/metabolism , Glucose Transporter Type 2/physiology , Homeodomain Proteins/physiology , Insulin/metabolism , Insulin-Secreting Cells/cytology , Mice , Mice, Transgenic , Models, Biological , Mutation , Time Factors , Trans-Activators/physiology , Transcription, Genetic
12.
Mol Cell Biol ; 26(15): 5735-43, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16847327

ABSTRACT

The MafA transcription factor is both critical to islet beta-cell function and has a unique pancreatic cell-type-specific expression pattern. To localize the potential transcriptional regulatory region(s) involved in directing expression to the beta cell, areas of identity within the 5' flanking region of the mouse, human, and rat mafA genes were found between nucleotides -9389 and -9194, -8426 and -8293, -8118 and -7750, -6622 and -6441, -6217 and -6031, and -250 and +56 relative to the transcription start site. The identity between species was greater than 75%, with the highest found between bp -8118 and -7750 ( approximately 94%, termed region 3). Region 3 was the only upstream mammalian conserved region found in chicken mafA (88% identity). In addition, region 3 uniquely displayed beta-cell-specific activity in cell-line-based reporter assays. Important regulators of beta-cell formation and function, PDX-1, FoxA2, and Nkx2.2, were shown to specifically bind to region 3 in vivo using the chromatin immunoprecipitation assay. Mutational and functional analyses demonstrated that FoxA2 (bp -7943 to -7910), Nkx2.2 (bp -7771 to -7746), and PDX-1 (bp -8087 to -8063) mediated region 3 activation. Consistent with a role in transcription, small interfering RNA-mediated knockdown of PDX-1 led to decreased mafA mRNA production in INS-1-derived beta-cell lines (832/13 and 832/3), while MafA expression was undetected in the pancreatic epithelium of Nkx2.2 null animals. These results suggest that beta-cell-type-specific mafA transcription is principally controlled by region 3-acting transcription factors that are essential in the formation of functional beta cells.


Subject(s)
Gene Expression Regulation , Hepatocyte Nuclear Factor 3-beta/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/physiology , Maf Transcription Factors, Large , Regulatory Sequences, Nucleic Acid , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Cell Line , Chickens , Hepatocyte Nuclear Factor 3-beta/genetics , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Humans , Insulin-Secreting Cells/cytology , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Mice , Molecular Sequence Data , Nuclear Proteins , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Sequence Alignment , Trans-Activators/genetics , Transcription Factors/genetics , Transcription, Genetic , Zebrafish Proteins
13.
Cell Metab ; 2(3): 153-63, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16154098

ABSTRACT

Diabetes causes pancreatic beta cell failure through hyperglycemia-induced oxidative stress, or "glucose toxicity." We show that the forkhead protein FoxO1 protects beta cells against oxidative stress by forming a complex with the promyelocytic leukemia protein Pml and the NAD-dependent deacetylase Sirt1 to activate expression of NeuroD and MafA, two Insulin2 (Ins2) gene transcription factors. Using acetylation-defective and acetylation-mimicking mutants, we demonstrate that acetylation targets FoxO1 to Pml and prevents ubiquitin-dependent degradation. We show that hyperglycemia suppresses MafA expression in vivo and that MafA inhibition can be prevented by transgenic expression of constitutively nuclear FoxO1 in beta cells. The findings provide a mechanism linking glucose- and growth factor receptor-activated pathways to protect beta cells against oxidative damage via FoxO proteins.


Subject(s)
Gene Expression Regulation , Islets of Langerhans/metabolism , Nerve Tissue Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Acetylation , Adenoviridae/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Diabetes Mellitus, Type 2/metabolism , Electrophoretic Mobility Shift Assay , Fluorescein-5-isothiocyanate , Fluorescent Antibody Technique , Fluorescent Dyes , Forkhead Box Protein O1 , Forkhead Transcription Factors , Hydrogen Peroxide/pharmacology , Immunohistochemistry , Luciferases/metabolism , Maf Transcription Factors, Large , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Point Mutation , Promoter Regions, Genetic , Promyelocytic Leukemia Protein , RNA, Messenger/metabolism , Sirtuin 1 , Sirtuins/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/metabolism , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL