Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37.245
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 32: 579-608, 2014.
Article in English | MEDLINE | ID: mdl-24499272

ABSTRACT

This paper reviews the presentation of peptides by major histocompatibility complex (MHC) class II molecules in the autoimmune diabetes of the nonobese diabetic (NOD) mouse. Islets of Langerhans contain antigen-presenting cells that capture the proteins and peptides of the beta cells' secretory granules. Peptides bound to I-A(g7), the unique MHC class II molecule of NOD mice, are presented in islets and in pancreatic lymph nodes. The various beta cell-derived peptides interact with selected CD4 T cells to cause inflammation and beta cell demise. Many autoreactive T cells are found in NOD mice, but not all have a major role in the initiation of the autoimmune process. I emphasize here the evidence pointing to insulin autoreactivity as a seminal component in the diabetogenic process.


Subject(s)
Antigen Presentation/immunology , Diabetes Mellitus, Type 1/immunology , Animals , Antigen-Presenting Cells/immunology , Autoantigens/immunology , Disease Models, Animal , Epitopes, T-Lymphocyte , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/immunology , Humans , Insulin/immunology , Islets of Langerhans/cytology , Islets of Langerhans/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Peptides/immunology , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocyte Subsets/immunology
2.
Cell ; 184(8): 1945-1948, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33831374

ABSTRACT

The 2021 Gairdner Prize is awarded to Daniel Drucker, Joel Habener, and Jens Juul Holst for the discovery of novel peptides encoded in the proglucagon sequence and the establishment of their physiological roles. These discoveries underpinned the development of therapeutics that are now benefiting patients with type 2 diabetes and other disorders worldwide.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide 1/therapeutic use , Glucagon-Like Peptide 2/therapeutic use , Proglucagon/chemistry , Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 2/chemistry , Glucagon-Like Peptide 2/metabolism , Humans , Islets of Langerhans/metabolism , Proglucagon/metabolism , Receptors, Glucagon/metabolism , Short Bowel Syndrome/drug therapy , Short Bowel Syndrome/metabolism
3.
Cell ; 180(6): 1198-1211.e19, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32200801

ABSTRACT

It has generally proven challenging to produce functional ß cells in vitro. Here, we describe a previously unidentified protein C receptor positive (Procr+) cell population in adult mouse pancreas through single-cell RNA sequencing (scRNA-seq). The cells reside in islets, do not express differentiation markers, and feature epithelial-to-mesenchymal transition characteristics. By genetic lineage tracing, Procr+ islet cells undergo clonal expansion and generate all four endocrine cell types during adult homeostasis. Sorted Procr+ cells, representing ∼1% of islet cells, can robustly form islet-like organoids when cultured at clonal density. Exponential expansion can be maintained over long periods by serial passaging, while differentiation can be induced at any time point in culture. ß cells dominate in differentiated islet organoids, while α, δ, and PP cells occur at lower frequencies. The organoids are glucose-responsive and insulin-secreting. Upon transplantation in diabetic mice, these organoids reverse disease. These findings demonstrate that the adult mouse pancreatic islet contains a population of Procr+ endocrine progenitors.


Subject(s)
Cell Culture Techniques/methods , Endothelial Protein C Receptor/metabolism , Islets of Langerhans/cytology , Animals , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Epithelial-Mesenchymal Transition/physiology , Female , Glucose/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Islets of Langerhans/growth & development , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Nude , Organoids/growth & development , Organoids/metabolism , Pancreas/cytology , Pancreas/metabolism , Protein C/metabolism , Stem Cells/cytology
4.
Cell ; 176(4): 790-804.e13, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30661759

ABSTRACT

The pancreatic islets of Langerhans regulate glucose homeostasis. The loss of insulin-producing ß cells within islets results in diabetes, and islet transplantation from cadaveric donors can cure the disease. In vitro production of whole islets, not just ß cells, will benefit from a better understanding of endocrine differentiation and islet morphogenesis. We used single-cell mRNA sequencing to obtain a detailed description of pancreatic islet development. Contrary to the prevailing dogma, we find islet morphology and endocrine differentiation to be directly related. As endocrine progenitors differentiate, they migrate in cohesion and form bud-like islet precursors, or "peninsulas" (literally "almost islands"). α cells, the first to develop, constitute the peninsular outer layer, and ß cells form later, beneath them. This spatiotemporal collinearity leads to the typical core-mantle architecture of the mature, spherical islet. Finally, we induce peninsula-like structures in differentiating human embryonic stem cells, laying the ground for the generation of entire islets in vitro.


Subject(s)
Islets of Langerhans/cytology , Islets of Langerhans/embryology , Animals , Cell Differentiation , Cells, Cultured , Human Embryonic Stem Cells/cytology , Humans , Insulin/metabolism , Insulin-Secreting Cells/cytology , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation/methods , Mice , Mice, Inbred C57BL , Mice, SCID , Morphogenesis , Pancreas/cytology
5.
Cell ; 168(5): 775-788.e12, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235195

ABSTRACT

Stem-cell-based therapies can potentially reverse organ dysfunction and diseases, but the removal of impaired tissue and activation of a program leading to organ regeneration pose major challenges. In mice, a 4-day fasting mimicking diet (FMD) induces a stepwise expression of Sox17 and Pdx-1, followed by Ngn3-driven generation of insulin-producing ß cells, resembling that observed during pancreatic development. FMD cycles restore insulin secretion and glucose homeostasis in both type 2 and type 1 diabetes mouse models. In human type 1 diabetes pancreatic islets, fasting conditions reduce PKA and mTOR activity and induce Sox2 and Ngn3 expression and insulin production. The effects of the FMD are reversed by IGF-1 treatment and recapitulated by PKA and mTOR inhibition. These results indicate that a FMD promotes the reprogramming of pancreatic cells to restore insulin generation in islets from T1D patients and reverse both T1D and T2D phenotypes in mouse models. PAPERCLIP.


Subject(s)
Diabetes Mellitus, Type 1/diet therapy , Diabetes Mellitus, Type 2/diet therapy , Fasting , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Diet , Glucose Tolerance Test , Humans , In Vitro Techniques , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans , Mice , Nerve Tissue Proteins/genetics , Pancreas/cytology , Pancreas/metabolism , Signal Transduction , Transcriptome
6.
Cell ; 168(1-2): 73-85.e11, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-27916274

ABSTRACT

The recent discovery that genetically modified α cells can regenerate and convert into ß-like cells in vivo holds great promise for diabetes research. However, to eventually translate these findings to human, it is crucial to discover compounds with similar activities. Herein, we report the identification of GABA as an inducer of α-to-ß-like cell conversion in vivo. This conversion induces α cell replacement mechanisms through the mobilization of duct-lining precursor cells that adopt an α cell identity prior to being converted into ß-like cells, solely upon sustained GABA exposure. Importantly, these neo-generated ß-like cells are functional and can repeatedly reverse chemically induced diabetes in vivo. Similarly, the treatment of transplanted human islets with GABA results in a loss of α cells and a concomitant increase in ß-like cell counts, suggestive of α-to-ß-like cell conversion processes also in humans. This newly discovered GABA-induced α cell-mediated ß-like cell neogenesis could therefore represent an unprecedented hope toward improved therapies for diabetes.


Subject(s)
Diabetes Mellitus/drug therapy , Glucagon-Secreting Cells/cytology , Insulin-Secreting Cells/cytology , gamma-Aminobutyric Acid/administration & dosage , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/drug effects , Diabetes Mellitus/chemically induced , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Glucagon-Secreting Cells/drug effects , Humans , Islets of Langerhans/cytology , Male , Mice , Nerve Tissue Proteins , Rats , Rats, Wistar , gamma-Aminobutyric Acid/pharmacology
7.
Cell ; 168(1-2): 86-100.e15, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-27916275

ABSTRACT

Type 1 diabetes is characterized by the destruction of pancreatic ß cells, and generating new insulin-producing cells from other cell types is a major aim of regenerative medicine. One promising approach is transdifferentiation of developmentally related pancreatic cell types, including glucagon-producing α cells. In a genetic model, loss of the master regulatory transcription factor Arx is sufficient to induce the conversion of α cells to functional ß-like cells. Here, we identify artemisinins as small molecules that functionally repress Arx by causing its translocation to the cytoplasm. We show that the protein gephyrin is the mammalian target of these antimalarial drugs and that the mechanism of action of these molecules depends on the enhancement of GABAA receptor signaling. Our results in zebrafish, rodents, and primary human pancreatic islets identify gephyrin as a druggable target for the regeneration of pancreatic ß cell mass from α cells.


Subject(s)
Artemisinins/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Disease Models, Animal , Receptors, GABA-A/metabolism , Signal Transduction , Animals , Artemether , Artemisinins/administration & dosage , Carrier Proteins/metabolism , Cell Transdifferentiation/drug effects , Cells, Cultured , Diabetes Mellitus/drug therapy , Diabetes Mellitus, Type 1/pathology , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Islets of Langerhans/drug effects , Membrane Proteins/metabolism , Mice , Protein Stability/drug effects , Rats , Single-Cell Analysis , Transcription Factors/metabolism , Zebrafish , gamma-Aminobutyric Acid/metabolism
8.
Nat Immunol ; 20(6): 677-686, 2019 06.
Article in English | MEDLINE | ID: mdl-31110312

ABSTRACT

Consumption of a high-energy Western diet triggers mild adaptive ß cell proliferation to compensate for peripheral insulin resistance; however, the underlying molecular mechanism remains unclear. In the present study we show that the toll-like receptors TLR2 and TLR4 inhibited the diet-induced replication of ß cells in mice and humans. The combined, but not the individual, loss of TLR2 and TLR4 increased the replication of ß cells, but not that of α cells, leading to enlarged ß cell area and hyperinsulinemia in diet-induced obesity. Loss of TLR2 and TLR4 increased the nuclear abundance of the cell cycle regulators cyclin D2 and Cdk4 in a manner dependent on the signaling mediator Erk. These data reveal a regulatory mechanism controlling the proliferation of ß cells in diet-induced obesity and suggest that selective targeting of the TLR2/TLR4 pathways may reverse ß cell failure in patients with diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Obesity/etiology , Obesity/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Animals , Cell Proliferation , Cyclin D2/metabolism , Cyclin-Dependent Kinase 4/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Humans , Insulin/blood , Insulin/metabolism , Insulin-Secreting Cells/ultrastructure , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Knockout , Multiprotein Complexes/metabolism , Obesity/drug therapy , Parabiosis , Protein Binding , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
9.
Genes Dev ; 37(11-12): 451-453, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37399332

ABSTRACT

The homeodomain transcription factor (TF) Nkx2.2 governs crucial cell fate decisions in several developing organs, including the central nervous system (CNS), pancreas, and intestine. How Nkx2.2 regulates unique targets in these different systems to impact their individual transcriptional programs remains unclear. In this issue of Genes & Development Abarinov and colleagues (pp. 490-504) generated and analyzed mice in which the Nkx2.2 SD is mutated and found that the SD is required for normal pancreatic islet differentiation but dispensable for most aspects of neuronal differentiation.


Subject(s)
Homeodomain Proteins , Islets of Langerhans , Mice , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeobox Protein Nkx-2.2 , Zebrafish Proteins/genetics , Islets of Langerhans/metabolism , Cell Differentiation/genetics , Neurons/metabolism , Gene Expression Regulation, Developmental
10.
Nature ; 627(8003): 347-357, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38374256

ABSTRACT

Type 2 diabetes (T2D) is a heterogeneous disease that develops through diverse pathophysiological processes1,2 and molecular mechanisms that are often specific to cell type3,4. Here, to characterize the genetic contribution to these processes across ancestry groups, we aggregate genome-wide association study data from 2,535,601 individuals (39.7% not of European ancestry), including 428,452 cases of T2D. We identify 1,289 independent association signals at genome-wide significance (P < 5 × 10-8) that map to 611 loci, of which 145 loci are, to our knowledge, previously unreported. We define eight non-overlapping clusters of T2D signals that are characterized by distinct profiles of cardiometabolic trait associations. These clusters are differentially enriched for cell-type-specific regions of open chromatin, including pancreatic islets, adipocytes, endothelial cells and enteroendocrine cells. We build cluster-specific partitioned polygenic scores5 in a further 279,552 individuals of diverse ancestry, including 30,288 cases of T2D, and test their association with T2D-related vascular outcomes. Cluster-specific partitioned polygenic scores are associated with coronary artery disease, peripheral artery disease and end-stage diabetic nephropathy across ancestry groups, highlighting the importance of obesity-related processes in the development of vascular outcomes. Our findings show the value of integrating multi-ancestry genome-wide association study data with single-cell epigenomics to disentangle the aetiological heterogeneity that drives the development and progression of T2D. This might offer a route to optimize global access to genetically informed diabetes care.


Subject(s)
Diabetes Mellitus, Type 2 , Disease Progression , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Adipocytes/metabolism , Chromatin/genetics , Chromatin/metabolism , Coronary Artery Disease/complications , Coronary Artery Disease/genetics , Diabetes Mellitus, Type 2/classification , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Nephropathies/complications , Diabetic Nephropathies/genetics , Endothelial Cells/metabolism , Enteroendocrine Cells , Epigenomics , Genetic Predisposition to Disease/genetics , Islets of Langerhans/metabolism , Multifactorial Inheritance/genetics , Peripheral Arterial Disease/complications , Peripheral Arterial Disease/genetics , Single-Cell Analysis
11.
Cell ; 159(4): 911-24, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25417165

ABSTRACT

The development of whole-body imaging at single-cell resolution enables system-level approaches to studying cellular circuits in organisms. Previous clearing methods focused on homogenizing mismatched refractive indices of individual tissues, enabling reductions in opacity but falling short of achieving transparency. Here, we show that an aminoalcohol decolorizes blood by efficiently eluting the heme chromophore from hemoglobin. Direct transcardial perfusion of an aminoalcohol-containing cocktail that we previously termed CUBIC coupled with a 10 day to 2 week clearing protocol decolorized and rendered nearly transparent almost all organs of adult mice as well as the entire body of infant and adult mice. This CUBIC-perfusion protocol enables rapid whole-body and whole-organ imaging at single-cell resolution by using light-sheet fluorescent microscopy. The CUBIC protocol is also applicable to 3D pathology, anatomy, and immunohistochemistry of various organs. These results suggest that whole-body imaging of colorless tissues at high resolution will contribute to organism-level systems biology.


Subject(s)
Amino Alcohols/analysis , Single-Cell Analysis/methods , Whole Body Imaging/methods , Animals , Diabetes Mellitus/pathology , Imaging, Three-Dimensional/methods , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL
12.
Cell ; 159(2): 428-39, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25303535

ABSTRACT

The generation of insulin-producing pancreatic ß cells from stem cells in vitro would provide an unprecedented cell source for drug discovery and cell transplantation therapy in diabetes. However, insulin-producing cells previously generated from human pluripotent stem cells (hPSC) lack many functional characteristics of bona fide ß cells. Here, we report a scalable differentiation protocol that can generate hundreds of millions of glucose-responsive ß cells from hPSC in vitro. These stem-cell-derived ß cells (SC-ß) express markers found in mature ß cells, flux Ca(2+) in response to glucose, package insulin into secretory granules, and secrete quantities of insulin comparable to adult ß cells in response to multiple sequential glucose challenges in vitro. Furthermore, these cells secrete human insulin into the serum of mice shortly after transplantation in a glucose-regulated manner, and transplantation of these cells ameliorates hyperglycemia in diabetic mice.


Subject(s)
Cell Culture Techniques , Insulin-Secreting Cells/cytology , Animals , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Insulin/genetics , Insulin/metabolism , Islets of Langerhans , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
13.
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
14.
Cell ; 158(3): 534-48, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25018104

ABSTRACT

Depending on endoplasmic reticulum (ER) stress levels, the ER transmembrane multidomain protein IRE1α promotes either adaptation or apoptosis. Unfolded ER proteins cause IRE1α lumenal domain homo-oligomerization, inducing trans autophosphorylation that further drives homo-oligomerization of its cytosolic kinase/endoribonuclease (RNase) domains to activate mRNA splicing of adaptive XBP1 transcription factor. However, under high/chronic ER stress, IRE1α surpasses an oligomerization threshold that expands RNase substrate repertoire to many ER-localized mRNAs, leading to apoptosis. To modulate these effects, we developed ATP-competitive IRE1α Kinase-Inhibiting RNase Attenuators-KIRAs-that allosterically inhibit IRE1α's RNase by breaking oligomers. One optimized KIRA, KIRA6, inhibits IRE1α in vivo and promotes cell survival under ER stress. Intravitreally, KIRA6 preserves photoreceptor functional viability in rat models of ER stress-induced retinal degeneration. Systemically, KIRA6 preserves pancreatic ß cells, increases insulin, and reduces hyperglycemia in Akita diabetic mice. Thus, IRE1α powerfully controls cell fate but can itself be controlled with small molecules to reduce cell degeneration.


Subject(s)
Endoplasmic Reticulum Stress , Endoribonucleases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Allosteric Regulation , Animals , Apoptosis/drug effects , Cell Line , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Enzyme Activation/drug effects , Humans , Islets of Langerhans/metabolism , Male , Mice , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Rats , Retina/metabolism , Ribonucleases/antagonists & inhibitors
15.
Nature ; 624(7992): 621-629, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38049589

ABSTRACT

Type 2 diabetes mellitus (T2D), a major cause of worldwide morbidity and mortality, is characterized by dysfunction of insulin-producing pancreatic islet ß cells1,2. T2D genome-wide association studies (GWAS) have identified hundreds of signals in non-coding and ß cell regulatory genomic regions, but deciphering their biological mechanisms remains challenging3-5. Here, to identify early disease-driving events, we performed traditional and multiplexed pancreatic tissue imaging, sorted-islet cell transcriptomics and islet functional analysis of early-stage T2D and control donors. By integrating diverse modalities, we show that early-stage T2D is characterized by ß cell-intrinsic defects that can be proportioned into gene regulatory modules with enrichment in signals of genetic risk. After identifying the ß cell hub gene and transcription factor RFX6 within one such module, we demonstrated multiple layers of genetic risk that converge on an RFX6-mediated network to reduce insulin secretion by ß cells. RFX6 perturbation in primary human islet cells alters ß cell chromatin architecture at regions enriched for T2D GWAS signals, and population-scale genetic analyses causally link genetically predicted reduced RFX6 expression with increased T2D risk. Understanding the molecular mechanisms of complex, systemic diseases necessitates integration of signals from multiple molecules, cells, organs and individuals, and thus we anticipate that this approach will be a useful template to identify and validate key regulatory networks and master hub genes for other diseases or traits using GWAS data.


Subject(s)
Diabetes Mellitus, Type 2 , Gene Expression Profiling , Gene Regulatory Networks , Genetic Predisposition to Disease , Islets of Langerhans , Humans , Case-Control Studies , Cell Separation , Chromatin/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Gene Regulatory Networks/genetics , Genome-Wide Association Study , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Reproducibility of Results
16.
Genes Dev ; 35(17-18): 1243-1255, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34385262

ABSTRACT

Multiple G protein-coupled receptors (GPCRs) are expressed in pancreatic islet cells, but the majority have unknown functions. We observed specific GPCRs localized to primary cilia, a prominent signaling organelle, in pancreatic α and ß cells. Loss of cilia disrupts ß-cell endocrine function, but the molecular drivers are unknown. Using functional expression, we identified multiple GPCRs localized to cilia in mouse and human islet α and ß cells, including FFAR4, PTGER4, ADRB2, KISS1R, and P2RY14. Free fatty acid receptor 4 (FFAR4) and prostaglandin E receptor 4 (PTGER4) agonists stimulate ciliary cAMP signaling and promote glucagon and insulin secretion by α- and ß-cell lines and by mouse and human islets. Transport of GPCRs to primary cilia requires TULP3, whose knockdown in primary human and mouse islets relocalized ciliary FFAR4 and PTGER4 and impaired regulated glucagon or insulin secretion, without affecting ciliary structure. Our findings provide index evidence that regulated hormone secretion by islet α and ß cells is controlled by ciliary GPCRs providing new targets for diabetes.


Subject(s)
Insulin-Secreting Cells , Islets of Langerhans , Animals , Glucagon/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Receptors, G-Protein-Coupled/genetics
17.
Immunity ; 50(1): 13-15, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30650372

ABSTRACT

The role of the immune system in homeostasis of pancreatic ß cells in type 2 diabetes mellitus is poorly characterized. In a recent issue of Cell Metabolism, Ying et al. (2018) report that two subpopulations of macrophages expand during obesity to impair ß cell function.


Subject(s)
Diabetes Mellitus, Type 2 , Islets of Langerhans , Cell Proliferation , Humans , Inflammation , Insulin , Macrophages , Obesity
18.
Cell ; 153(2): 413-25, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23582329

ABSTRACT

Here, we demonstrate that the fractalkine (FKN)/CX3CR1 system represents a regulatory mechanism for pancreatic islet ß cell function and insulin secretion. CX3CR1 knockout (KO) mice exhibited a marked defect in glucose and GLP1-stimulated insulin secretion, and this defect was also observed in vitro in isolated islets from CX3CR1 KO mice. In vivo administration of FKN improved glucose tolerance with an increase in insulin secretion. In vitro treatment of islets with FKN increased intracellular Ca(2+) and potentiated insulin secretion in both mouse and human islets. The KO islets exhibited reduced expression of a set of genes necessary for the fully functional, differentiated ß cell state, whereas treatment of wild-type (WT) islets with FKN led to increased expression of these genes. Lastly, expression of FKN in islets was decreased by aging and high-fat diet/obesity, suggesting that decreased FKN/CX3CR1 signaling could be a mechanism underlying ß cell dysfunction in type 2 diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , Receptors, Chemokine/metabolism , Signal Transduction , Adult , Aging , Animals , CX3C Chemokine Receptor 1 , Cadaver , Chemokine CX3CL1/administration & dosage , Chemokine CX3CL1/metabolism , Diet, High-Fat , Gene Expression , Glucose/metabolism , Humans , Hyperglycemia/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Receptors, Chemokine/genetics
19.
Genes Dev ; 34(15-16): 1089-1105, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32616519

ABSTRACT

The circadian clock is encoded by a negative transcriptional feedback loop that coordinates physiology and behavior through molecular programs that remain incompletely understood. Here, we reveal rhythmic genome-wide alternative splicing (AS) of pre-mRNAs encoding regulators of peptidergic secretion within pancreatic ß cells that are perturbed in Clock-/- and Bmal1-/- ß-cell lines. We show that the RNA-binding protein THRAP3 (thyroid hormone receptor-associated protein 3) regulates circadian clock-dependent AS by binding to exons at coding sequences flanking exons that are more frequently skipped in clock mutant ß cells, including transcripts encoding Cask (calcium/calmodulin-dependent serine protein kinase) and Madd (MAP kinase-activating death domain). Depletion of THRAP3 restores expression of the long isoforms of Cask and Madd, and mimicking exon skipping in these transcripts through antisense oligonucleotide delivery in wild-type islets reduces glucose-stimulated insulin secretion. Finally, we identify shared networks of alternatively spliced exocytic genes from islets of rodent models of diet-induced obesity that significantly overlap with clock mutants. Our results establish a role for pre-mRNA alternative splicing in ß-cell function across the sleep/wake cycle.


Subject(s)
Alternative Splicing , Circadian Clocks/genetics , Exocytosis , Glucose/metabolism , Insulin Secretion/genetics , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/physiology , Animals , CLOCK Proteins/genetics , CLOCK Proteins/physiology , Cells, Cultured , Death Domain Receptor Signaling Adaptor Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Homeostasis , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Male , Mice, Inbred C57BL , Nuclear Proteins/physiology , Obesity/genetics , Obesity/metabolism , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism , Transcription Factors/physiology
20.
Development ; 151(2)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38265192

ABSTRACT

The autonomic nervous system innervates the pancreas by sympathetic, parasympathetic and sensory branches during early organogenesis, starting with neural crest cell invasion and formation of an intrinsic neuronal network. Several studies have demonstrated that signals from pancreatic neural crest cells direct pancreatic endocrinogenesis. Likewise, autonomic neurons have been shown to regulate pancreatic islet formation, and have also been implicated in type I diabetes. Here, we provide an overview of recent progress in mapping pancreatic innervation and understanding the interactions between pancreatic neurons, epithelial morphogenesis and cell differentiation. Finally, we discuss pancreas innervation as a factor in the development of diabetes.


Subject(s)
Diabetes Mellitus , Islets of Langerhans , Humans , Cell Differentiation , Organogenesis , Pancreas
SELECTION OF CITATIONS
SEARCH DETAIL