Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Cell ; 157(7): 1577-90, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24949970

ABSTRACT

Clec16a has been identified as a disease susceptibility gene for type 1 diabetes, multiple sclerosis, and adrenal dysfunction, but its function is unknown. Here we report that Clec16a is a membrane-associated endosomal protein that interacts with E3 ubiquitin ligase Nrdp1. Loss of Clec16a leads to an increase in the Nrdp1 target Parkin, a master regulator of mitophagy. Islets from mice with pancreas-specific deletion of Clec16a have abnormal mitochondria with reduced oxygen consumption and ATP concentration, both of which are required for normal ß cell function. Indeed, pancreatic Clec16a is required for normal glucose-stimulated insulin release. Moreover, patients harboring a diabetogenic SNP in the Clec16a gene have reduced islet Clec16a expression and reduced insulin secretion. Thus, Clec16a controls ß cell function and prevents diabetes by controlling mitophagy. This pathway could be targeted for prevention and control of diabetes and may extend to the pathogenesis of other Clec16a- and Parkin-associated diseases.


Subject(s)
Diabetes Mellitus, Type 1/genetics , Islets of Langerhans/pathology , Lectins, C-Type/metabolism , Mitophagy , Monosaccharide Transport Proteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Diabetes Mellitus, Type 1/pathology , Genetic Predisposition to Disease , Glucose/metabolism , Humans , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Lysosomes/chemistry , Lysosomes/metabolism , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/genetics , Polymorphism, Single Nucleotide , Ubiquitin-Protein Ligases
2.
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
3.
Proc Natl Acad Sci U S A ; 114(6): 1341-1346, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28115692

ABSTRACT

The stress response and cell survival are necessary for normal pancreatic ß-cell function, glucose homeostasis, and prevention of diabetes. The homeodomain transcription factor and human diabetes gene pancreas/duodenum homeobox protein 1 (Pdx1) regulates ß-cell survival and endoplasmic reticulum stress susceptibility, in part through direct regulation of activating transcription factor 4 (Atf4). Here we show that Atf5, a close but less-studied relative of Atf4, is also a target of Pdx1 and is critical for ß-cell survival under stress conditions. Pdx1 deficiency led to decreased Atf5 transcript, and primary islet ChIP-sequencing localized PDX1 to the Atf5 promoter, implicating Atf5 as a PDX1 target. Atf5 expression was stress inducible and enriched in ß cells. Importantly, Atf5 deficiency decreased survival under stress conditions. Loss-of-function and chromatin occupancy experiments positioned Atf5 downstream of and parallel to Atf4 in the regulation of eIF4E-binding protein 1 (4ebp1), a mammalian target of rapamycin (mTOR) pathway component that inhibits protein translation. Accordingly, Atf5 deficiency attenuated stress suppression of global translation, likely enhancing the susceptibility of ß cells to stress-induced apoptosis. Thus, we identify ATF5 as a member of the transcriptional network governing pancreatic ß-cell survival during stress.


Subject(s)
Activating Transcription Factors/genetics , Apoptosis/genetics , Endoplasmic Reticulum Stress/genetics , Insulin-Secreting Cells/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Activating Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cells, Cultured , Eukaryotic Initiation Factors , Gene Expression Regulation , Gene Regulatory Networks , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
4.
Am J Physiol Endocrinol Metab ; 314(4): E308-E321, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29351489

ABSTRACT

The transcription factors pancreatic and duodenal homeobox 1 (Pdx1) and onecut1 (Oc1) are coexpressed in multipotent pancreatic progenitors (MPCs), but their expression patterns diverge in hormone-expressing cells, with Oc1 expression being extinguished in the endocrine lineage and Pdx1 being maintained at high levels in ß-cells. We previously demonstrated that cooperative function of these two factors in MPCs is necessary for proper specification and differentiation of pancreatic endocrine cells. In those studies, we observed a persistent decrease in expression of the ß-cell maturity factor MafA. We therefore hypothesized that Pdx1 and Oc1 cooperativity in MPCs impacts postnatal ß-cell maturation and function. Here our model of Pdx1-Oc1 double heterozygosity was used to investigate the impact of haploinsufficiency for both of these factors on postnatal ß-cell maturation, function, and adaptability. Examining mice at postnatal day (P) 14, we observed alterations in pancreatic insulin content in both Pdx1 heterozygotes and double heterozygotes. Gene expression analysis at this age revealed significantly decreased expression of many genes important for glucose-stimulated insulin secretion (e.g., Glut2, Pcsk1/2, Abcc8) exclusively in double heterozygotes. Analysis of P14 islets revealed an increase in the number of mixed islets in double heterozygotes. We predicted that double-heterozygous ß-cells would have an impaired ability to respond to stress. Indeed, we observed that ß-cell proliferation fails to increase in double heterozygotes in response to either high-fat diet or placental lactogen. We thus report here the importance of cooperation between regulatory factors early in development for postnatal islet maturation and adaptability.


Subject(s)
Hepatocyte Nuclear Factor 6/physiology , Homeodomain Proteins/physiology , Insulin-Secreting Cells/physiology , Islets of Langerhans/growth & development , Multipotent Stem Cells/metabolism , Trans-Activators/physiology , Adaptation, Physiological/drug effects , Adaptation, Physiological/genetics , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Diet, High-Fat , Gene Expression Regulation, Developmental/drug effects , Glucose/pharmacology , Hepatocyte Nuclear Factor 6/genetics , Homeodomain Proteins/genetics , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/physiology , Male , Mice , Mice, Transgenic , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/physiology , Organogenesis/drug effects , Organogenesis/genetics , Trans-Activators/genetics
5.
Physiol Genomics ; 49(2): 105-114, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28011883

ABSTRACT

The heterogeneity of the developing pancreatic epithelium and low abundance of endocrine progenitors limit the information derived from traditional expression studies. To identify genes that characterize early developmental tissues composed of multiple progenitor lineages, we applied single-cell RNA-Seq to embryonic day (e)13.5 mouse pancreata and performed integrative analysis with single cell data from mature pancreas. We identified subpopulations expressing macrophage or endothelial markers and new pancreatic progenitor markers. We also identified potential α-cell precursors expressing glucagon (Gcg) among the e13.5 pancreatic cells. Despite their high Gcg expression levels, these cells shared greater transcriptomic similarity with other e13.5 cells than with adult α-cells, indicating their immaturity. Comparative analysis identified the sodium-dependent neutral amino acid transporter, Slc38a5, as a characteristic gene expressed in α-cell precursors but not mature cells. By immunofluorescence analysis, we observed SLC38A5 expression in pancreatic progenitors, including in a subset of NEUROG3+ endocrine progenitors and MAFB+ cells and in all GCG+ cells. Expression declined in α-cells during late gestation and was absent in the adult islet. Our results suggest SLC38A5 as an early marker of α-cell lineage commitment.


Subject(s)
Gene Expression Profiling , Pancreas/cytology , Single-Cell Analysis , Stem Cells/metabolism , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Cell Differentiation/genetics , Cluster Analysis , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Islets of Langerhans/cytology , Mice , Models, Biological , Stem Cells/cytology
6.
Am J Physiol Endocrinol Metab ; 313(6): E651-E662, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28811293

ABSTRACT

Pharmacological activation of the glucagon-like peptide-1 receptor (GLP-1R) in the ventromedial hypothalamus (VMH) reduces food intake. Here, we assessed whether suppression of food intake by GLP-1R agonists (GLP-1RA) in this region is dependent on AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR). We found that pharmacological inhibition of glycolysis, and thus activation of AMPK, in the VMH attenuates the anorectic effect of the GLP-1R agonist exendin-4 (Ex4), indicating that glucose metabolism and inhibition of AMPK are both required for this effect. Furthermore, we found that Ex4-mediated anorexia in the VMH involved mTOR but not acetyl-CoA carboxylase, two downstream targets of AMPK. We support this by showing that Ex4 activates mTOR signaling in the VMH and Chinese hamster ovary (CHO)-K1 cells. In contrast to the clear acute pharmacological impact of the these receptors on food intake, knockdown of the VMH Glp1r conferred no changes in energy balance in either chow- or high-fat-diet-fed mice, and the acute anorectic and glucose tolerance effects of peripherally dosed GLP-1RA were preserved. These results show that the VMH GLP-1R regulates food intake by engaging key nutrient sensors but is dispensable for the effects of GLP-1RA on nutrient homeostasis.


Subject(s)
Eating/physiology , Food , Glucagon-Like Peptide-1 Receptor/physiology , Sensation/physiology , Ventromedial Hypothalamic Nucleus/physiology , Acetyl-CoA Carboxylase/metabolism , Adenylate Kinase/metabolism , Animals , Body Composition/drug effects , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Eating/drug effects , Exenatide , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Glycolysis/drug effects , Homeostasis/physiology , Male , Mice , Mice, Inbred C57BL , Peptides/pharmacology , Sensation/drug effects , TOR Serine-Threonine Kinases/metabolism , Venoms/pharmacology , Ventromedial Hypothalamic Nucleus/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
9.
J Clin Invest ; 134(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38950317

ABSTRACT

Glucose plays a key role in shaping pancreatic ß cell function. Thus, deciphering the mechanisms by which this nutrient stimulates ß cells holds therapeutic promise for combating ß cell failure in type 2 diabetes (T2D). ß Cells respond to hyperglycemia in part by rewiring their mRNA metabolism, yet the mechanisms governing these changes remain poorly understood. Here, we identify a requirement for the RNA-binding protein PCBP2 in maintaining ß cell function basally and during sustained hyperglycemic challenge. PCBP2 was induced in primary mouse islets incubated with elevated glucose and was required to adapt insulin secretion. Transcriptomic analysis of primary Pcbp2-deficient ß cells revealed impacts on basal and glucose-regulated mRNAs encoding core components of the insulin secretory pathway. Accordingly, Pcbp2-deficient ß cells exhibited defects in calcium flux, insulin granule ultrastructure and exocytosis, and the amplification pathway of insulin secretion. Further, PCBP2 was induced by glucose in primary human islets, was downregulated in islets from T2D donors, and impacted genes commonly altered in islets from donors with T2D and linked to single-nucleotide polymorphisms associated with T2D. Thus, these findings establish a paradigm for PCBP2 in governing basal and glucose-adaptive gene programs critical for shaping the functional state of ß cells.


Subject(s)
Diabetes Mellitus, Type 2 , Glucose , Insulin-Secreting Cells , Insulin , RNA-Binding Proteins , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Animals , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Mice , Humans , Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Insulin/metabolism , Insulin Secretion , Mice, Knockout , Male , Adaptation, Physiological
10.
Diabetes ; 73(4): 554-564, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38266068

ABSTRACT

Assessment of pancreas cell type composition is crucial to the understanding of the genesis of diabetes. Current approaches use immunodetection of protein markers, for example, insulin as a marker of ß-cells. A major limitation of these methods is that protein content varies in physiological and pathological conditions, complicating the extrapolation to actual cell number. Here, we demonstrate the use of cell type-specific DNA methylation markers for determining the fraction of specific cell types in human islet and pancreas specimens. We identified genomic loci that are uniquely demethylated in specific pancreatic cell types and applied targeted PCR to assess the methylation status of these loci in tissue samples, enabling inference of cell type composition. In islet preparations, normalization of insulin secretion to ß-cell DNA revealed similar ß-cell function in pre-type 1 diabetes (T1D), T1D, and type 2 diabetes (T2D), which was significantly lower than in donors without diabetes. In histological pancreas specimens from recent-onset T1D, this assay showed ß-cell fraction within the normal range, suggesting a significant contribution of ß-cell dysfunction. In T2D pancreata, we observed increased α-cell fraction and normal ß-cell fraction. Methylation-based analysis provides an accurate molecular alternative to immune detection of cell types in the human pancreas, with utility in the interpretation of insulin secretion assays and the assessment of pancreas cell composition in health and disease.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Glucagon-Secreting Cells , Insulin-Secreting Cells , Islets of Langerhans , Humans , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Islets of Langerhans/metabolism , DNA Methylation , Pancreas/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Glucagon-Secreting Cells/metabolism
11.
Proc Natl Acad Sci U S A ; 107(47): 20494-9, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21059917

ABSTRACT

Three-amino-acid-loop-extension (TALE) homeodomain proteins including Meis and Pbx families are generally recognized for their roles in growth and differentiation during vertebrate embryogenesis and tumorigenesis. Whereas genetic studies indicate that Pbx1 regulates the development and function of insulin-producing pancreatic ß-cells, the role of Meis family members in ß-cells is still unknown. Here we show that Meis3 is abundantly expressed in pancreatic islets and ß-cells and that it regulates ß-cell survival. We further identify the 3-phosphoinositide-dependent protein kinase 1 (PDK1), a well-known kinase involved in the PI3K-Akt signaling pathway, as a direct Meis3 target, which mediates its role in ß-cell survival. This regulatory module appears to function broadly as we also identify Meis3 regulation of cell survival and PDK1 expression in ovarian carcinoma cells, suggesting a unique function for Meis3 beyond the traditional roles for TALE homeodomain factors during embryogenesis.


Subject(s)
Apoptosis/genetics , Homeodomain Proteins/metabolism , Islets of Langerhans/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/physiology , Chromatin Immunoprecipitation , DNA Primers/genetics , Homeodomain Proteins/genetics , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Microarray Analysis , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
12.
Proc Natl Acad Sci U S A ; 107(47): 20358-63, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21059956

ABSTRACT

A hallmark of diabetes is an absolute or relative reduction in the number of functional ß cells. Therapies that could increase the number of endogenous ß cells under diabetic conditions would be desirable. Prevalent gene targeting mouse models for assessing ß-cell proliferation and diabetes pathogenesis only address whether deletion of a gene prevents the development of diabetes. Models testing whether acute excision of a single gene can ameliorate or reverse preexisting hyperglycemia in established diabetes remain to be explored, which could directly validate the effect of gene excision on treating diabetes. Here, we report that acute and temporally controlled excision of the Men1 gene, which encodes menin, ameliorated preexisting hyperglycemia in streptozotocin-treated mice. Moreover, Men1 excision also improved the preexisting hyperglycemia and glucose intolerance in genetic db/db diabetic mice. Furthermore, acute Men1 excision reversed preexisting glucose intolerance in high-fat diet-fed mice. Men1 excision improved glucose metabolism at least partly through increasing proliferation of endogenous ß cells and islet size. Acute Men1 excision up-regulated a group of proproliferative genes in pancreatic islets. Together, these findings demonstrate that established hyperglycemia can be reversed through repression of a single gene, Men1, in diabetic conditions, and suggest that menin is a vital regulator in pathogenesis of diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Gene Deletion , Gene Expression Regulation/physiology , Hyperglycemia/genetics , Insulin-Secreting Cells/cytology , Proto-Oncogene Proteins/genetics , Animals , Blotting, Western , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Glucose Tolerance Test , Immunohistochemistry , Insulin/blood , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
13.
Proc Natl Acad Sci U S A ; 106(45): 19090-5, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19855005

ABSTRACT

Type 2 diabetes mellitus (T2DM) results from pancreatic beta cell failure in the setting of insulin resistance. Heterozygous mutations in the gene encoding the beta cell transcription factor pancreatic duodenal homeobox 1 (Pdx1) are associated with both T2DM and maturity onset diabetes of the young (MODY4), and low levels of Pdx1 accompany beta cell dysfunction in experimental models of glucotoxicity and diabetes. Here, we find that Pdx1 is required for compensatory beta cell mass expansion in response to diet-induced insulin resistance through its roles in promoting beta cell survival and compensatory hypertrophy. Pdx1-deficient beta cells show evidence of endoplasmic reticulum (ER) stress both in the complex metabolic milieu of high-fat feeding as well as in the setting of acutely reduced Pdx1 expression in the Min6 mouse insulinoma cell line. Further, Pdx1 deficiency enhances beta cell susceptibility to ER stress-associated apoptosis. The results of high throughput expression microarray and chromatin occupancy analyses reveal that Pdx1 regulates a broad array of genes involved in diverse functions of the ER, including proper disulfide bond formation, protein folding, and the unfolded protein response. These findings suggest that Pdx1 deficiency leads to a failure of beta cell compensation for insulin resistance at least in part by impairing critical functions of the ER.


Subject(s)
Apoptosis/genetics , Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression Regulation/genetics , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Trans-Activators/metabolism , Animals , Cell Enlargement , Cell Line, Tumor , Chromatin Immunoprecipitation , Gene Silencing , Homeodomain Proteins/genetics , Insulin Resistance/physiology , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Trans-Activators/genetics
14.
J Clin Invest ; 132(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35642629

ABSTRACT

BACKGROUNDMultiple islet autoantibodies (AAbs) predict the development of type 1 diabetes (T1D) and hyperglycemia within 10 years. By contrast, T1D develops in only approximately 15% of individuals who are positive for single AAbs (generally against glutamic acid decarboxylase [GADA]); hence, the single GADA+ state may represent an early stage of T1D.METHODSHere, we functionally, histologically, and molecularly phenotyped human islets from nondiabetic GADA+ and T1D donors.RESULTSSimilar to the few remaining ß cells in the T1D islets, GADA+ donor islets demonstrated a preserved insulin secretory response. By contrast, α cell glucagon secretion was dysregulated in both GADA+ and T1D islets, with impaired glucose suppression of glucagon secretion. Single-cell RNA-Seq of GADA+ α cells revealed distinct abnormalities in glycolysis and oxidative phosphorylation pathways and a marked downregulation of cAMP-dependent protein kinase inhibitor ß (PKIB), providing a molecular basis for the loss of glucose suppression and the increased effect of 3-isobutyl-1-methylxanthine (IBMX) observed in GADA+ donor islets.CONCLUSIONWe found that α cell dysfunction was present during the early stages of islet autoimmunity at a time when ß cell mass was still normal, raising important questions about the role of early α cell dysfunction in the progression of T1D.FUNDINGThis work was supported by grants from the NIH (3UC4DK112217-01S1, U01DK123594-02, UC4DK112217, UC4DK112232, U01DK123716, and P30 DK019525) and the Vanderbilt Diabetes Research and Training Center (DK20593).


Subject(s)
Diabetes Mellitus, Type 1 , Glutamate Decarboxylase , Autoantibodies , Glucagon , Glucose , Humans
15.
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
16.
J Clin Invest ; 118(6): 2316-24, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18464933

ABSTRACT

Intrauterine growth retardation (IUGR) has been linked to the onset of diseases in adulthood, including type 2 diabetes, and has been proposed to result from altered gene regulation patterns due to epigenetic modifications of developmental genes. To determine whether epigenetic modifications may play a role in the development of adult diabetes following IUGR, we used a rodent model of IUGR that expresses lower levels of Pdx1, a pancreatic and duodenal homeobox 1 transcription factor critical for beta cell function and development, which develops diabetes in adulthood. We found that expression of Pdx1 was permanently reduced in IUGR beta cells and underwent epigenetic modifications throughout development. The fetal IUGR state was characterized by loss of USF-1 binding at the proximal promoter of Pdx1, recruitment of the histone deacetylase 1 (HDAC1) and the corepressor Sin3A, and deacetylation of histones H3 and H4. Following birth, histone 3 lysine 4 (H3K4) was demethylated and histone 3 lysine 9 (H3K9) was methylated. During the neonatal period, these epigenetic changes and the reduction in Pdx1 expression could be reversed by HDAC inhibition. After the onset of diabetes in adulthood, the CpG island in the proximal promoter was methylated, resulting in permanent silencing of the Pdx1 locus. These results provide insight into the development of type 2 diabetes following IUGR and we believe they are the first to describe the ontogeny of chromatin remodeling in vivo from the fetus to the onset of disease in adulthood.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Epigenesis, Genetic , Fetal Growth Retardation/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Animals , Chromatin/chemistry , CpG Islands , DNA Methylation , Histones/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism
17.
Proc Natl Acad Sci U S A ; 105(48): 18913-8, 2008 Dec 02.
Article in English | MEDLINE | ID: mdl-19028870

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is believed to arise through a multistep model comprised of putative precursor lesions known as pancreatic intraepithelial neoplasia (PanIN). Recent genetically engineered mouse models of PDAC demonstrate a comparable morphologic spectrum of murine PanIN (mPanIN) lesions. The histogenesis of PanIN and PDAC in both mice and men remains controversial. The most faithful genetic models activate an oncogenic Kras(G12D) knockin allele within the pdx1- or ptf1a/p48-expression domain of the entire pancreatic anlage during development, thus obscuring the putative cell(s)-of-origin from which subsequent mPanIN lesions arise. In our study, activation of this knockin Kras(G12D) allele in the Elastase- and Mist1-expressing mature acinar compartment of adult mice resulted in the spontaneous induction of mPanIN lesions of all histological grades, although invasive carcinomas per se were not seen. We observed no requirement for concomitant chronic exocrine injury in the induction of mPanIN lesions from the mature acinar cell compartment. The acinar cell derivation of the mPanINs was established through lineage tracing in reporter mice, and by microdissection of lesional tissue demonstrating Cre-mediated recombination events. In contrast to the uniformly penetrant mPanIN phenotype observed following developmental activation of Kras(G12D) in the Pdx1-expressing progenitor cells, the Pdx1-expressing population in the mature pancreas (predominantly islet beta cells) appears to be relatively resistant to the effects of oncogenic Kras. We conclude that in the appropriate genetic context, the differentiated acinar cell compartment in adult mice retains its susceptibility for spontaneous transformation into mPanIN lesions, a finding with potential relevance vis-à-vis the origins of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Pancreas, Exocrine/cytology , Pancreas, Exocrine/metabolism , Pancreatic Neoplasms/metabolism , Precancerous Conditions/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Carcinoma, Pancreatic Ductal/pathology , Humans , Mice , Mice, Transgenic , Pancreas, Exocrine/pathology , Pancreatic Neoplasms/pathology , Precancerous Conditions/pathology , Promoter Regions, Genetic , Proto-Oncogene Proteins p21(ras)/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/physiology
18.
Diabetes ; 70(4): 903-916, 2021 04.
Article in English | MEDLINE | ID: mdl-33526589

ABSTRACT

Current evidence indicates that proliferating ß-cells express lower levels of some functional cell identity genes, suggesting that proliferating cells are not optimally functional. Pdx1 is important for ß-cell specification, function, and proliferation and is mutated in monogenic forms of diabetes. However, its regulation during the cell cycle is unknown. Here we examined Pdx1 protein expression in immortalized ß-cells, maternal mouse islets during pregnancy, and mouse embryonic pancreas. We demonstrate that Pdx1 localization and protein levels are highly dynamic. In nonmitotic cells, Pdx1 is not observed in constitutive heterochromatin, nucleoli, or most areas containing repressive epigenetic marks. At prophase, Pdx1 is enriched around the chromosomes before Ki67 coating of the chromosome surface. Pdx1 uniformly localizes in the cytoplasm at prometaphase and becomes enriched around the chromosomes again at the end of cell division, before nuclear envelope formation. Cells in S phase have lower Pdx1 levels than cells at earlier cell cycle stages, and overexpression of Pdx1 in INS-1 cells prevents progression toward G2, suggesting that cell cycle-dependent regulation of Pdx1 is required for completion of mitosis. Together, we find that Pdx1 localization and protein levels are tightly regulated throughout the cell cycle. This dynamic regulation has implications for the dichotomous role of Pdx1 in ß-cell function and proliferation.


Subject(s)
Cell Cycle/physiology , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/physiology , Trans-Activators/metabolism , Animals , Blotting, Western , Cell Cycle/genetics , Cell Line , Cell Proliferation/genetics , Cell Proliferation/physiology , Female , Flow Cytometry , Gene Expression Regulation , Homeodomain Proteins/genetics , Humans , Insulin-Secreting Cells/metabolism , Ki-67 Antigen/metabolism , Mice , Trans-Activators/genetics
19.
J Clin Invest ; 117(4): 971-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17404620

ABSTRACT

It has been suggested that pancreatic acinar cells can serve as progenitors for pancreatic islets, a concept with substantial implications for therapeutic efforts to increase insulin-producing beta cell mass in patients with diabetes. We report what we believe to be the first in vivo lineage tracing approach to determine the plasticity potential of pancreatic acinar cells. We developed an acinar cell-specific inducible Cre recombinase transgenic mouse, which, when mated with a reporter strain and pulsed with tamoxifen, resulted in permanent and specific labeling of acinar cells and their progeny. During various time periods of observation and using several models to provoke injury, we failed to observe any chase of the labeled cells into the endocrine compartment, indicating that acinar cells do not normally transdifferentiate into islet beta cells in vivo in adult mice. In contrast, we observed a substantial role for replication of preexisting acinar cells in the regeneration of new acinar cells after partial pancreatectomy. These results indicate that mature acinar cells harbor a facultative acinar but not endocrine progenitor capacity.


Subject(s)
Insulin-Secreting Cells/physiology , Integrases/genetics , Pancreas/cytology , Pancreas/physiology , Animals , Digestive System Physiological Phenomena , Insulin-Secreting Cells/drug effects , Integrases/metabolism , Mice , Mice, Transgenic , Organ Specificity , Pancreas/drug effects , Rats , Regeneration , Reverse Transcriptase Polymerase Chain Reaction , Tamoxifen/pharmacology
20.
Diabetes ; 69(4): 499-507, 2020 04.
Article in English | MEDLINE | ID: mdl-32198193

ABSTRACT

In type 2 diabetes, ß-cells endure various forms of cellular stress, including oxidative stress and endoplasmic reticulum stress, secondary to increased demand for insulin production and extracellular perturbations, including hyperglycemia. Chronic exposure to stress causes impaired insulin secretion, apoptosis, and loss of cell identity, and a combination of these processes leads to ß-cell failure and severe hyperglycemia. Therefore, a better understanding of the molecular mechanisms underlying stress responses in ß-cells promises to reveal new therapeutic opportunities for type 2 diabetes. In this perspective, we discuss posttranscriptional control of gene expression as a critical, but underappreciated, layer of regulation with broad importance during stress responses. Specifically, regulation of mRNA translation occurs pervasively during stress to activate gene expression programs; however, the convenience of RNA sequencing has caused translational regulation to be overlooked compared with transcriptional controls. We highlight the role of RNA binding proteins in shaping selective translational regulation during stress and the mechanisms underlying this level of regulation. A growing body of evidence indicates that RNA binding proteins control an array of processes in ß-cells, including the synthesis and secretion of insulin. Therefore, systematic evaluations of translational regulation and the upstream factors shaping this level of regulation are critical areas of investigation to expand our understanding of ß-cell failure in type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Endoplasmic Reticulum Stress/physiology , Insulin-Secreting Cells/metabolism , Oxidative Stress/physiology , RNA Processing, Post-Transcriptional/physiology , RNA-Binding Proteins/metabolism , Animals , Apoptosis/physiology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Gene Expression Regulation , Humans , Insulin Secretion/physiology , Insulin-Secreting Cells/pathology , Protein Processing, Post-Translational/physiology , RNA-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL