1
|
Perez-Frances M, Bru-Tari E, Cohrs C, Abate MV, van Gurp L, Furuyama K, Speier S, Thorel F, Herrera PL. Regulated and adaptive in vivo insulin secretion from islets only containing β-cells. Nat Metab 2024; 6:1791-1806. [PMID: 39169271 PMCID: PMC11422169 DOI: 10.1038/s42255-024-01114-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/22/2024] [Indexed: 08/23/2024]
Abstract
Insulin-producing β-cells in pancreatic islets are regulated by systemic cues and, locally, by adjacent islet hormone-producing 'non-β-cells' (namely α-cells, δ-cells and γ-cells). Yet whether the non-β-cells are required for accurate insulin secretion is unclear. Here, we studied mice in which adult islets are exclusively composed of β-cells and human pseudoislets containing only primary β-cells. Mice lacking non-β-cells had optimal blood glucose regulation, enhanced glucose tolerance, insulin sensitivity and restricted body weight gain under a high-fat diet. The insulin secretion dynamics in islets composed of only β-cells was comparable to that in intact islets. Similarly, human β-cell pseudoislets retained the glucose-regulated mitochondrial respiration, insulin secretion and exendin-4 responses of entire islets. The findings indicate that non-β-cells are dispensable for blood glucose homeostasis and β-cell function. These results support efforts aimed at developing diabetes treatments by generating β-like clusters devoid of non-β-cells, such as from pluripotent stem cells differentiated in vitro or by reprograming non-β-cells into insulin producers in situ.
Collapse
Affiliation(s)
- Marta Perez-Frances
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Eva Bru-Tari
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christian Cohrs
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
| | - Maria Valentina Abate
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Léon van Gurp
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Kenichiro Furuyama
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Stephan Speier
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
| | - Fabrizio Thorel
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pedro L Herrera
- Department of Genetic Medicine and Development, iGE3 and Centre facultaire du diabète, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
2
|
Perrelli M, Goparaju P, Postolache TT, del Bosque-Plata L, Gragnoli C. Stress and the CRH System, Norepinephrine, Depression, and Type 2 Diabetes. Biomedicines 2024; 12:1187. [PMID: 38927393 PMCID: PMC11200886 DOI: 10.3390/biomedicines12061187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Major depressive disorder (MDD) increases the risk of type 2 diabetes (T2D) by 60% in untreated patients, and hypercortisolism is common in MDD as well as in some patients with T2D. Patients with MDD, despite hypercortisolism, show inappropriately normal levels of corticotropin-releasing hormone (CRH) and plasma adrenocorticotropin (ACTH) in the cerebrospinal fluid, which might implicate impaired negative feedback. Also, a positive feedback loop of the CRH-norepinephrine (NE)-CRH system may be involved in the hypercortisolism of MDD and T2D. Dysfunctional CRH receptor 1 (CRHR1) and CRH receptor 2 (CRHR2), both of which are involved in glucose regulation, may explain hypercortisolism in MDD and T2D, at least in a subgroup of patients. CRHR1 increases glucose-stimulated insulin secretion. Dysfunctional CRHR1 variants can cause hypercortisolism, leading to serotonin dysfunction and depression, which can contribute to hyperglycemia, insulin resistance, and increased visceral fat, all of which are characteristics of T2D. CRHR2 is implicated in glucose homeostasis through the regulation of insulin secretion and gastrointestinal functions, and it stimulates insulin sensitivity at the muscular level. A few studies show a correlation of the CRHR2 gene with depressive disorders. Based on our own research, we have found a linkage and association (i.e., linkage disequilibrium [LD]) of the genes CRHR1 and CRHR2 with MDD and T2D in families with T2D. The correlation of CRHR1 and CRHR2 with MDD appears stronger than that with T2D, and per our hypothesis, MDD may precede the onset of T2D. According to the findings of our analysis, CRHR1 and CRHR2 variants could modify the response to prolonged chronic stress and contribute to high levels of cortisol, increasing the risk of developing MDD, T2D, and the comorbidity MDD-T2D. We report here the potential links of the CRH system, NE, and their roles in MDD and T2D.
Collapse
Affiliation(s)
| | - Pruthvi Goparaju
- Division of Endocrinology, Department of Medicine, Creighton University School of Medicine, Omaha, NE 68124, USA;
| | - Teodor T. Postolache
- Mood and Anxiety Program, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 19, Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO 80246, USA
- Mental Illness Research Education and Clinical Center (MIRECC), Veterans Integrated Service Network (VISN) 5, VA Capitol Health Care Network, Baltimore, MD 21090, USA
| | - Laura del Bosque-Plata
- Nutrigenetics, and Nutrigenomic Laboratory, National Institute of Genomic Medicine, Mexico City 14610, Mexico;
| | - Claudia Gragnoli
- Division of Endocrinology, Department of Medicine, Creighton University School of Medicine, Omaha, NE 68124, USA;
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA 17033, USA
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, 8091 Zürich, Switzerland
- Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, 00197 Rome, Italy
| |
Collapse
|
3
|
Devère M, Takhlidjt S, Prévost G, Chartrel N, Leprince J, Picot M. The 26RFa (QRFP)/GPR103 Neuropeptidergic System: A Key Regulator of Energy and Glucose Metabolism. Neuroendocrinology 2024:1-17. [PMID: 38599200 DOI: 10.1159/000538629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/28/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Obesity and type 2 diabetes are strongly associated pathologies, currently considered as a worldwide epidemic problem. Understanding the mechanisms that drive the development of these diseases would enable to develop new therapeutic strategies for their prevention and treatment. Particularly, the role of the brain in energy and glucose homeostasis has been studied for 2 decades. In specific, the hypothalamus contains well-identified neural networks that regulate appetite and potentially also glucose homeostasis. A new concept has thus emerged, suggesting that obesity and diabetes could be due to a dysfunction of the same, still poorly understood, neural networks. SUMMARY The neuropeptide 26RFa (also termed QRFP) belongs to the family of RFamide regulatory peptides and has been identified as the endogenous ligand of the human G protein-coupled receptor GPR103 (QRFPR). The primary structure of 26RFa is strongly conserved during vertebrate evolution, suggesting its crucial roles in the control of vital functions. Indeed, the 26RFa/GPR103 peptidergic system is reported to be involved in the control of various neuroendocrine functions, notably the control of energy metabolism in which it plays an important role, both centrally and peripherally, since 26RFa regulates feeding behavior, thermogenesis and lipogenesis. Moreover, 26RFa is reported to control glucose homeostasis both peripherally, where it acts as an incretin, and centrally, where the 26RFa/GPR103 system relays insulin signaling in the brain to control glucose metabolism. KEY MESSAGES This review gives a comprehensive overview of the role of the 26RFa/GPR103 system as a key player in the control of energy and glucose metabolism. In a pathophysiological context, this neuropeptidergic system represents a prime therapeutic target whose mechanisms are highly relevant to decipher.
Collapse
Affiliation(s)
- Mélodie Devère
- University Rouen Normandie, Inserm, NorDiC UMR 1239, Normandie University, Rouen, France
| | - Saloua Takhlidjt
- University Rouen Normandie, Inserm, NorDiC UMR 1239, Normandie University, Rouen, France
| | - Gaëtan Prévost
- University Rouen Normandie, Inserm, NorDiC UMR 1239, Normandie University, Rouen, France
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Rouen Normandie, Inserm, Normandie University, NorDiC UMR 1239, CHU Rouen, Rouen, France
| | - Nicolas Chartrel
- University Rouen Normandie, Inserm, NorDiC UMR 1239, Normandie University, Rouen, France
| | - Jérôme Leprince
- University Rouen Normandie, Inserm, NorDiC UMR 1239, Normandie University, Rouen, France
- University Rouen Normandie, Normandie University, INSERM US 51, CNRS UAR 2026, HeRacLeS, Rouen, France
| | - Marie Picot
- University Rouen Normandie, Inserm, NorDiC UMR 1239, Normandie University, Rouen, France
| |
Collapse
|
4
|
Hill TG, Hill DJ. The Importance of Intra-Islet Communication in the Function and Plasticity of the Islets of Langerhans during Health and Diabetes. Int J Mol Sci 2024; 25:4070. [PMID: 38612880 PMCID: PMC11012451 DOI: 10.3390/ijms25074070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Islets of Langerhans are anatomically dispersed within the pancreas and exhibit regulatory coordination between islets in response to nutritional and inflammatory stimuli. However, within individual islets, there is also multi-faceted coordination of function between individual beta-cells, and between beta-cells and other endocrine and vascular cell types. This is mediated partly through circulatory feedback of the major secreted hormones, insulin and glucagon, but also by autocrine and paracrine actions within the islet by a range of other secreted products, including somatostatin, urocortin 3, serotonin, glucagon-like peptide-1, acetylcholine, and ghrelin. Their availability can be modulated within the islet by pericyte-mediated regulation of microvascular blood flow. Within the islet, both endocrine progenitor cells and the ability of endocrine cells to trans-differentiate between phenotypes can alter endocrine cell mass to adapt to changed metabolic circumstances, regulated by the within-islet trophic environment. Optimal islet function is precariously balanced due to the high metabolic rate required by beta-cells to synthesize and secrete insulin, and they are susceptible to oxidative and endoplasmic reticular stress in the face of high metabolic demand. Resulting changes in paracrine dynamics within the islets can contribute to the emergence of Types 1, 2 and gestational diabetes.
Collapse
Affiliation(s)
- Thomas G. Hill
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - David J. Hill
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON N6A 4V2, Canada;
- Departments of Medicine, Physiology and Pharmacology, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
5
|
Amin M, Horst N, Wu R, Gragnoli C. Novel corticotropin-releasing hormone receptor genes (CRHR1 and CRHR2) linkage to and association with polycystic ovary syndrome. J Ovarian Res 2023; 16:155. [PMID: 37543650 PMCID: PMC10403835 DOI: 10.1186/s13048-023-01159-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 04/10/2023] [Indexed: 08/07/2023] Open
Abstract
BACKGROUND Women with polycystic ovarian syndrome (PCOS) have increased hypothalamic-pituitary-adrenal (HPA) axis activation, pro-inflammatory mediators, and psychological distress in response to stressors. In women with PCOS, the corticotropin-releasing hormone (CRH) induces an exaggerated HPA response, possibly mediated by one of the CRH receptors (CRHR1 or CRHR2). Both CRHR1 and CRHR2 are implicated in insulin secretion, and variants in CRHR1 and CRHR2 genes may predispose to the mental-metabolic risk for PCOS. METHODS We phenotyped 212 Italian families with type 2 diabetes (T2D) for PCOS following the Rotterdam diagnostic criteria. We analyzed within CRHR1 and CRHR2 genes, respectively, 36 and 18 microarray-variants for parametric linkage to and/or linkage disequilibrium (LD) with PCOS under the recessive with complete penetrance (R1) and dominant with complete penetrance (D1) models. Subsequentially, we ran a secondary analysis under the models dominant with incomplete penetrance (D2) and recessive with incomplete penetrance (R2). RESULTS We detected 22 variants in CRHR1 and 1 variant in CRHR2 significantly (p < 0.05) linked to or in LD with PCOS across different inheritance models. CONCLUSIONS This is the first study to report CRHR1 and CRHR2 as novel risk genes in PCOS. In silico analysis predicted that the detected CRHR1 and CRHR2 risk variants promote negative chromatin activation of their related genes in the ovaries, potentially affecting the female cycle and ovulation. However, CRHR1- and CRHR2-risk variants might also lead to hypercortisolism and confer mental-metabolic pleiotropic effects. Functional studies are needed to confirm the pathogenicity of genes and related variants.
Collapse
Affiliation(s)
- Mutaz Amin
- INSERM, US14-Orphanet, 75014, Paris, France
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Al-Neelain University, Khartoum, Sudan, 11121
| | - Nicholas Horst
- Division of Endocrinology, Department of Medicine, Creighton University School of Medicine, Omaha, NE, 68124, USA
| | - Rongling Wu
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, 17033, USA
- Department of Statistics, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Claudia Gragnoli
- Division of Endocrinology, Department of Medicine, Creighton University School of Medicine, Omaha, NE, 68124, USA.
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, 17033, USA.
- Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, 00197, Rome, Italy.
| |
Collapse
|
6
|
Mawla AM, van der Meulen T, Huising MO. Chromatin accessibility differences between alpha, beta, and delta cells identifies common and cell type-specific enhancers. BMC Genomics 2023; 24:202. [PMID: 37069576 PMCID: PMC10108528 DOI: 10.1186/s12864-023-09293-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 04/03/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND High throughput sequencing has enabled the interrogation of the transcriptomic landscape of glucagon-secreting alpha cells, insulin-secreting beta cells, and somatostatin-secreting delta cells. These approaches have furthered our understanding of expression patterns that define healthy or diseased islet cell types and helped explicate some of the intricacies between major islet cell crosstalk and glucose regulation. All three endocrine cell types derive from a common pancreatic progenitor, yet alpha and beta cells have partially opposing functions, and delta cells modulate and control insulin and glucagon release. While gene expression signatures that define and maintain cellular identity have been widely explored, the underlying epigenetic components are incompletely characterized and understood. However, chromatin accessibility and remodeling is a dynamic attribute that plays a critical role to determine and maintain cellular identity. RESULTS Here, we compare and contrast the chromatin landscape between mouse alpha, beta, and delta cells using ATAC-Seq to evaluate the significant differences in chromatin accessibility. The similarities and differences in chromatin accessibility between these related islet endocrine cells help define their fate in support of their distinct functional roles. We identify patterns that suggest that both alpha and delta cells are poised, but repressed, from becoming beta-like. We also identify patterns in differentially enriched chromatin that have transcription factor motifs preferentially associated with different regions of the genome. Finally, we not only confirm and visualize previously discovered common endocrine- and cell specific- enhancer regions across differentially enriched chromatin, but identify novel regions as well. We compiled our chromatin accessibility data in a freely accessible database of common endocrine- and cell specific-enhancer regions that can be navigated with minimal bioinformatics expertise. CONCLUSIONS Both alpha and delta cells appear poised, but repressed, from becoming beta cells in murine pancreatic islets. These data broadly support earlier findings on the plasticity in identity of non-beta cells under certain circumstances. Furthermore, differential chromatin accessibility shows preferentially enriched distal-intergenic regions in beta cells, when compared to either alpha or delta cells.
Collapse
Affiliation(s)
- Alex M Mawla
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Talitha van der Meulen
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, One Shields Avenue, Davis, CA, 95616, USA
| | - Mark O Huising
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, One Shields Avenue, Davis, CA, 95616, USA.
- Department of Physiology and Membrane Biology, School of Medicine, University of California, One Shields Avenue, Davis, CA, 95616, USA.
| |
Collapse
|
7
|
Corticotropin-Releasing Hormone: Biology and Therapeutic Opportunities. BIOLOGY 2022; 11:biology11121785. [PMID: 36552294 PMCID: PMC9775501 DOI: 10.3390/biology11121785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/16/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
In 1981, Wylie Vale, Joachim Spiess, Catherine Rivier, and Jean Rivier reported on the characterization of a 41-amino-acid peptide from ovine hypothalamic extracts with high potency and intrinsic activity stimulating the secretion of adrenocorticotropic hormone and β-endorphin by cultured anterior pituitary cells. With its sequence known, this neuropeptide was determined to be a hormone and consequently named corticotropin-releasing hormone (CRH), although the term corticotropin-releasing factor (CRF) is still used and preferred in some circumstances. Several decades have passed since this seminal contribution that opened a new research era, expanding the understanding of the coding of stress-related processes. The characterization of CRH receptors, the availability of CRH agonists and antagonists, and advanced immunocytochemical staining techniques have provided evidence that CRH plays a role in the regulation of several biological systems. The purpose of this review is to summarize the present knowledge of this 41-amino-acid peptide.
Collapse
|
8
|
Eskandari F, Salimi M, Binayi F, Abdollahifar MA, Eftekhary M, Hedayati M, Ghanbarian H, Zardooz H. Investigating the Effects of Maternal Separation on Hypothalamic-Pituitary-Adrenal Axis and Glucose Homeostasis under Chronic Social Defeat Stress in Young Adult Male Rat Offspring. Neuroendocrinology 2022; 113:361-380. [PMID: 36088912 DOI: 10.1159/000526989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Given the suggested metabolic regulatory effects of stress-responsive genes and based on the impacts of early-life stress on HPA axis development, this study aimed to characterize the maternal separation (MS) impact on the communication between glucose metabolism and HPA axis dysregulations under chronic social defeat stress (CSDS). METHODS During the first 2 weeks of life, male Wistar rats were either exposed to MS or left undisturbed with their mothers (Std). Starting on postnatal day 50, the animals of each group were either left undisturbed in the standard group housing (Con) or underwent CSDS for 3 weeks. There were four groups (n = 10/group): Std-Con, MS-Con, Std-CSDS, and MS-CSDS. RESULTS Early and/or adult life adversity reduced β-cell number, muscular FK506-binding protein 51 (FKBP51) content, and BMI in adulthood. The reduction of β-cell number and BMI in the MS-CSDS rats were more profound than MS-Con group. CSDS either alone or in combination with MS reduced locomotor activity and increased and decreased corticotropin-releasing factor type 1 receptor (CRFR1) content, respectively, in hypothalamus and pancreas. Although, under CSDS, MS intensified HPA axis overactivity and reduced isolated islets' insulin secretion, it could promote resilience to depression symptoms. No differences were observed in hypothalamic Fkbp5 gene DNA methylation and glucose tolerance among groups. CONCLUSION MS exacerbated HPA axis overactivity and the endocrine pancreas dysfunctions under CSDS. The intensified corticosterone secretion and the diminished content of pancreatic CRFR1 protein could be involved in the reduced β-cell number and islets' insulin secretion under CSDS. The decreased muscular FKBP51 content might be a homeostatic response to slow down insulin resistance development under chronic stress.
Collapse
Affiliation(s)
- Farzaneh Eskandari
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Salimi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fateme Binayi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohamad Eftekhary
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbarian
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Mao Y, Schoenborn J, Wang Z, Chen X, Matson K, Mohan R, Zhang S, Tang X, Arunagiri A, Arvan P, Tang X. Transgenic overexpression of microRNA-30d in pancreatic beta-cells progressively regulates beta-cell function and identity. Sci Rep 2022; 12:11969. [PMID: 35831364 PMCID: PMC9279310 DOI: 10.1038/s41598-022-16174-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 07/06/2022] [Indexed: 11/15/2022] Open
Abstract
Abnormal microRNA functions are closely associated with pancreatic β-cell loss and dysfunction in type 2 diabetes. Dysregulation of miR-30d has been reported in the individuals with diabetes. To study how miR-30d affects pancreatic β-cell functions, we generated two transgenic mouse lines that specifically overexpressed miR-30d in β-cells at distinct low and high levels. Transgenic overexpressed miR-30d systemically affected β-cell function. Elevated miR-30d at low-level (TgL, 2-fold) had mild effects on signaling pathways and displayed no significant changes to metabolic homeostasis. In contrast, transgenic mice with high-level of miR-30d expression (TgH, 12-fold) exhibited significant diet-induced hyperglycemia and β-cell dysfunction. In addition, loss of β-cell identity was invariably accompanied with increased insulin/glucagon-double positive bihormonal cells and excess plasma glucagon levels. The transcriptomic analysis revealed that miR-30d overexpression inhibited β-cell-enriched gene expression and induced α-cell-enriched gene expression. These findings implicate that an appropriate miR-30d level is essential in maintaining normal β-cell identity and function.
Collapse
Affiliation(s)
- Yiping Mao
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, 49931, USA
| | - Jacob Schoenborn
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, 49931, USA
| | - Zhihong Wang
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, 49931, USA
| | - Xinqian Chen
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, 49931, USA
| | - Katy Matson
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, 49931, USA
| | - Ramkumar Mohan
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, 49931, USA
| | - Shungang Zhang
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, 49931, USA
| | - Xiaohu Tang
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, 49931, USA
| | - Anoop Arunagiri
- Department of Metabolism Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Peter Arvan
- Department of Metabolism Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Xiaoqing Tang
- Department of Biological Sciences, Michigan Technological University, Houghton, MI, 49931, USA.
| |
Collapse
|
10
|
Flisher MF, Shin D, Huising MO. Urocortin3: Local inducer of somatostatin release and bellwether of beta cell maturity. Peptides 2022; 151:170748. [PMID: 35065098 PMCID: PMC10881066 DOI: 10.1016/j.peptides.2022.170748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/03/2022] [Accepted: 01/17/2022] [Indexed: 11/25/2022]
Abstract
Urocortin 3 (UCN3) is a peptide hormone expressed in pancreatic islets of Langerhans of both human alpha and human beta cells and solely in murine beta cells. UCN3 signaling acts locally within the islet to activate its cognate receptor, corticotropin releasing hormone receptor 2 (CRHR2), which is expressed by delta cells, to potentiate somatostatin (SST) negative feedback to reduce islet cell hormone output. The functional importance of UCN3 signaling in the islet is to modulate the amount of SST tone allowing for finely tuned regulation of insulin and glucagon secretion. UCN3 signaling is a hallmark of functional beta cell maturation, increasing the beta cell glucose threshold for insulin secretion. In doing so, UCN3 plays a relevant functional role in accurately maintaining blood glucose homeostasis. Additionally, UCN3 acts as an indicator of beta cell maturation and health, as UCN3 is not expressed in immature beta cells and is downregulated in dedifferentiated and dysfunctional beta cell states. Here, we review the mechanistic underpinnings of UCN3 signaling, its net effect on islet cell hormone output, as well as its value as a marker for beta cell maturation and functional status.
Collapse
Affiliation(s)
- Marcus F Flisher
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, Davis, CA, United States
| | - Donghan Shin
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, Davis, CA, United States
| | - Mark O Huising
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, Davis, CA, United States; Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, United States.
| |
Collapse
|
11
|
Khan D, Moffett RC, Flatt PR, Tarasov AI. Classical and non-classical islet peptides in the control of β-cell function. Peptides 2022; 150:170715. [PMID: 34958851 DOI: 10.1016/j.peptides.2021.170715] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/25/2021] [Accepted: 12/17/2021] [Indexed: 12/25/2022]
Abstract
The dual role of the pancreas as both an endocrine and exocrine gland is vital for food digestion and control of nutrient metabolism. The exocrine pancreas secretes enzymes into the small intestine aiding digestion of sugars and fats, whereas the endocrine pancreas secretes a cocktail of hormones into the blood, which is responsible for blood glucose control and regulation of carbohydrate, protein and fat metabolism. Classical islet hormones, insulin, glucagon, pancreatic polypeptide and somatostatin, interact in an autocrine and paracrine manner, to fine-tube the islet function and insulin secretion to the needs of the body. Recently pancreatic islets have been reported to express a number of non-classical peptide hormones involved in metabolic signalling, whose major production site was believed to reside outside pancreas, e.g. in the small intestine. We highlight the key non-classical islet peptides, and consider their involvement, together with established islet hormones, in regulation of stimulus-secretion coupling as well as proliferation, survival and transdifferentiation of β-cells. We furthermore focus on the paracrine interaction between classical and non-classical islet hormones in the maintenance of β-cell function. Understanding the functional relationships between these islet peptides might help to develop novel, more efficient treatments for diabetes and related metabolic disorders.
Collapse
Affiliation(s)
- Dawood Khan
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK.
| | - R Charlotte Moffett
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Peter R Flatt
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| | - Andrei I Tarasov
- Biomedical Sciences Research Institute, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland, UK
| |
Collapse
|
12
|
Pretorius M, Huang C. Beta-Cell Adaptation to Pregnancy - Role of Calcium Dynamics. Front Endocrinol (Lausanne) 2022; 13:853876. [PMID: 35399944 PMCID: PMC8990731 DOI: 10.3389/fendo.2022.853876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/21/2022] [Indexed: 11/17/2022] Open
Abstract
During pregnancy, the mother develops insulin resistance to shunt nutrients to the growing fetus. As a result, the maternal islets of Langerhans undergo several changes to increase insulin secretion in order to maintain glucose homeostasis and prevent the development of gestational diabetes. These changes include an increase in β-cell proliferation and β-cell mass, upregulation of insulin synthesis and insulin content, enhanced cell-to-cell communication, and a lowering of the glucose threshold for insulin secretion, all of which resulting in an increase in glucose-stimulated insulin secretion. Emerging data suggests that a change in intracellular calcium dynamics occurs in the β-cell during pregnancy as part of the adaptive process. Influx of calcium into β-cells is crucial in the regulation of glucose-stimulated insulin secretion. Calcium fluxes into and out of the cytosol, endoplasmic reticulum, and mitochondria are also important in controlling β-cell function and survival. Here, we review calcium dynamics in islets in response to pregnancy-induced changes in hormones and signaling molecules, and how these changes may enhance insulin secretion to stave off gestational diabetes.
Collapse
|
13
|
Abstract
This review focuses on the human pancreatic islet-including its structure, cell composition, development, function, and dysfunction. After providing a historical timeline of key discoveries about human islets over the past century, we describe new research approaches and technologies that are being used to study human islets and how these are providing insight into human islet physiology and pathophysiology. We also describe changes or adaptations in human islets in response to physiologic challenges such as pregnancy, aging, and insulin resistance and discuss islet changes in human diabetes of many forms. We outline current and future interventions being developed to protect, restore, or replace human islets. The review also highlights unresolved questions about human islets and proposes areas where additional research on human islets is needed.
Collapse
Affiliation(s)
- John T Walker
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Diane C Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marcela Brissova
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
14
|
Koike S, Hsu MF, Bettaieb A, Chu B, Matsumoto N, Morisseau C, Havel PJ, Huising MO, Hammock BD, Haj FG. Genetic deficiency or pharmacological inhibition of soluble epoxide hydrolase ameliorates high fat diet-induced pancreatic β-cell dysfunction and loss. Free Radic Biol Med 2021; 172:48-57. [PMID: 34038767 PMCID: PMC9901526 DOI: 10.1016/j.freeradbiomed.2021.05.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic β-cells are crucial regulators of systemic glucose homeostasis, and their dysfunction and loss are central features in type 2 diabetes. Interventions that rectify β-cell dysfunction and loss are essential to combat this deadly malady. In the current study, we sought to delineate the role of soluble epoxide hydrolase (sEH) in β-cells under diet-induced metabolic stress. The expression of sEH was upregulated in murine and macaque diabetes models and islets of diabetic human patients. We postulated that hyperglycemia-induced elevation in sEH leads to a reduction in its substrates, epoxyeicosatrienoic acids (EETs), and attenuates the function of β-cells. Genetic deficiency of sEH potentiated glucose-stimulated insulin secretion in mice, likely in a cell-autonomous manner, contributing to better systemic glucose control. Consistent with this observation, genetic and pharmacological inactivation of sEH and the treatment with EETs exhibited insulinotropic effects in isolated murine islets ex vivo. Additionally, sEH deficiency enhanced glucose sensing and metabolism with elevated ATP and cAMP concentrations. This phenotype was associated with attenuated oxidative stress and diminished β-cell death in sEH deficient islets. Moreover, pharmacological inhibition of sEH in vivo mitigated, albeit partly, high fat diet-induced β-cell loss and dedifferentiation. The current observations provide new insights into the role of sEH in β-cells and information that may be leveraged for the development of a mechanism-based intervention to rectify β-cell dysfunction and loss.
Collapse
Affiliation(s)
- Shinichiro Koike
- Department of Nutrition, University of California Davis, Davis, CA, 95616, USA
| | - Ming-Fo Hsu
- Department of Nutrition, University of California Davis, Davis, CA, 95616, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of California Davis, Davis, CA, 95616, USA
| | - Bryan Chu
- Department of Nutrition, University of California Davis, Davis, CA, 95616, USA
| | - Naoki Matsumoto
- Department of Entomology and Nematology, University of California Davis, Davis, CA, 95616, USA
| | - Christophe Morisseau
- Department of Entomology and Nematology, University of California Davis, Davis, CA, 95616, USA; Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA
| | - Peter J Havel
- Department of Nutrition, University of California Davis, Davis, CA, 95616, USA; Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA, 95616, USA
| | - Mark O Huising
- Department of Neurobiology & Physiology and Behavior, University of California Davis, Davis, CA, 95616, USA; Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, CA, 95616, USA
| | - Bruce D Hammock
- Department of Entomology and Nematology, University of California Davis, Davis, CA, 95616, USA; Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA
| | - Fawaz G Haj
- Department of Nutrition, University of California Davis, Davis, CA, 95616, USA; Comprehensive Cancer Center, University of California Davis, Sacramento, CA, 95817, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, University of California Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
15
|
Abstract
Pancreatic beta cells are the only cell type in our body capable of producing and secreting insulin to instruct the insulin-sensitive cells and tissues of our bodies to absorb nutrients after a meal. Accurate control of insulin release is of critical importance; too little insulin leads to diabetes, while an excess of insulin can cause potentially fatal hypoglycaemia. Yet, the pancreas of most people will control insulin secretion safely and effectively over decades and in response to glucose excursions driven by tens of thousands of meals. Because we only become aware of the important contributions of the pancreas when it fails to maintain glucose homeostasis, it is easy to forget just how well insulin release from a healthy pancreas is matched to insulin need to ensure stable blood glucose levels. Beta cells achieve this feat by extensive crosstalk with the rest of the endocrine cell types in the islet, notably the glucagon-producing alpha cells and somatostatin-producing delta cells. Here I will review the important paracrine contributions that each of these cells makes to the stimulation and subsequent inhibition of insulin release in response to a transient nutrient stimulation, and make the case that a breakdown of this local crosstalk contributes to the pathophysiology of diabetes. Graphical abstract.
Collapse
Affiliation(s)
- Mark O Huising
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, 196 Briggs Hall, 1 Shields Avenue, Davis, CA, 95616, USA.
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA.
| |
Collapse
|
16
|
Discovery of a stable tripeptide targeting the N-domain of CRF1 receptor. Amino Acids 2020; 52:1337-1351. [PMID: 32996057 DOI: 10.1007/s00726-020-02895-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 09/19/2020] [Indexed: 10/23/2022]
Abstract
The corticotropin-releasing factor (CRF) and its CRF1 receptor (CRF1R) play a central role in the maintenance of homeostasis. Malfunctioning of the CRF/CRF1R unit is associated with several disorders, such as anxiety and depression. Non-peptide CRF1R-selective antagonists have been shown to exert anxiolytic and antidepressant effects on experimental animals. However, none of them is in clinical use today because of several side effects, thus demonstrating the need for the development of other more suitable CRF1R antagonists. In an effort to develop novel CRF1R antagonists we designed, synthesized and chemically characterized two tripeptide analogues of CRF, namely (R)-LMI and (S)-LMI, having their Leu either in R (or D) or in S (or L) configuration, respectively. Their design was based on the crystal structure of the N-extracellular domain (N-domain) of CRF1R/CRF complex, using a relevant array of computational methods. Experimental evaluation of the stability of synthetic peptides in human plasma has revealed that (R)-LMI is proteolytically more stable than (S)-LMI. Based on this finding, (R)-LMI was selected for pharmacological characterization. We have found that (R)-LMI is a CRF antagonist, inhibiting (1) the CRF-stimulated accumulation of cAMP in HEK 293 cells expressing the CRF1R, (2) the production of interleukins by adipocytes and (3) the proliferation rate of RAW 264.7 cells. (R)-LMI likely blocked agonist actions by interacting with the N-domain of CRF1R as suggested by data using a constitutively active chimera of CRF1R. We propose that (R)-LMI can be used as an optimal lead compound in the rational design of novel CRF antagonists.
Collapse
|
17
|
Simpson SJS, Smith LIF, Jones PM, Bowe JE. UCN2: a new candidate influencing pancreatic β-cell adaptations in pregnancy. J Endocrinol 2020; 245:247-257. [PMID: 32106091 PMCID: PMC7159164 DOI: 10.1530/joe-19-0568] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 02/27/2020] [Indexed: 12/11/2022]
Abstract
The corticotropin-releasing hormone (CRH) family of peptides, including urocortin (UCN) 1, 2 and 3, are established hypothalamic neuroendocrine peptides, regulating the physiological and behaviour responses to stress indirectly, via the hypothalamic-pituitary-adrenal (HPA) axis. More recently, these peptides have been implicated in diverse roles in peripheral organs through direct signalling, including in placental and pancreatic islet physiology. CRH has been shown to stimulate insulin release through activation of its cognate receptors, CRH receptor 1 (CRHR1) and 2. However, the physiological significance of this is unknown. We have previously reported that during mouse pregnancy, expression of CRH peptides increase in mouse placenta suggesting that these peptides may play a role in various biological functions associated with pregnancy, particularly the pancreatic islet adaptations that occur in the pregnant state to compensate for the physiological increase in maternal insulin resistance. In the current study, we show that mouse pregnancy is associated with increased circulating levels of UCN2 and that when we pharmacologically block endogenous CRHR signalling in pregnant mice, impairment of glucose tolerance is observed. This effect on glucose tolerance was comparable to that displayed with specific CRHR2 blockade and not with specific CRHR1 blockade. No effects on insulin sensitivity or the proliferative capacity of β-cells were detected. Thus, CRHR2 signalling appears to be involved in β-cell adaptive responses to pregnancy in the mouse, with endogenous placental UCN2 being the likely signal mediating this.
Collapse
Affiliation(s)
- Sian J S Simpson
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Science and Medicine, King’s College London, London, UK
- Correspondence should be addressed to S J S Simpson:
| | - Lorna I F Smith
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Science and Medicine, King’s College London, London, UK
| | - Peter M Jones
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Science and Medicine, King’s College London, London, UK
| | - James E Bowe
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Science and Medicine, King’s College London, London, UK
| |
Collapse
|
18
|
El-Mehdi M, Takhlidjt S, Khiar F, Prévost G, do Rego JL, do Rego JC, Benani A, Nedelec E, Godefroy D, Arabo A, Lefranc B, Leprince J, Anouar Y, Chartrel N, Picot M. Glucose homeostasis is impaired in mice deficient in the neuropeptide 26RFa (QRFP). BMJ Open Diabetes Res Care 2020; 8:8/1/e000942. [PMID: 32114486 PMCID: PMC7050347 DOI: 10.1136/bmjdrc-2019-000942] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/09/2020] [Accepted: 01/28/2020] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION 26RFa (pyroglutamyl RFamide peptide (QRFP)) is a biologically active peptide that has been found to control feeding behavior by stimulating food intake, and to regulate glucose homeostasis by acting as an incretin. The aim of the present study was thus to investigate the impact of 26RFa gene knockout on the regulation of energy and glucose metabolism. RESEARCH DESIGN AND METHODS 26RFa mutant mice were generated by homologous recombination, in which the entire coding region of prepro26RFa was replaced by the iCre sequence. Energy and glucose metabolism was evaluated through measurement of complementary parameters. Morphological and physiological alterations of the pancreatic islets were also investigated. RESULTS Our data do not reveal significant alteration of energy metabolism in the 26RFa-deficient mice except the occurrence of an increased basal metabolic rate. By contrast, 26RFa mutant mice exhibited an altered glycemic phenotype with an increased hyperglycemia after a glucose challenge associated with an impaired insulin production, and an elevated hepatic glucose production. Two-dimensional and three-dimensional immunohistochemical experiments indicate that the insulin content of pancreatic β cells is much lower in the 26RFa-/- mice as compared with the wild-type littermates. CONCLUSION Disruption of the 26RFa gene induces substantial alteration in the regulation of glucose homeostasis, with in particular a deficit in insulin production by the pancreatic islets. These findings further support the notion that 26RFa is an important regulator of glucose homeostasis.
Collapse
|
19
|
Yip L, Fuhlbrigge R, Alkhataybeh R, Fathman CG. Gene Expression Analysis of the Pre-Diabetic Pancreas to Identify Pathogenic Mechanisms and Biomarkers of Type 1 Diabetes. Front Endocrinol (Lausanne) 2020; 11:609271. [PMID: 33424774 PMCID: PMC7793767 DOI: 10.3389/fendo.2020.609271] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/16/2020] [Indexed: 12/28/2022] Open
Abstract
Type 1 Diabetes (T1D) occurs as a result of the autoimmune destruction of pancreatic β-cells by self-reactive T cells. The etiology of this disease is complex and difficult to study due to a lack of disease-relevant tissues from pre-diabetic individuals. In this study, we performed gene expression analysis on human pancreas tissues obtained from the Network of Pancreatic Organ Donors with Diabetes (nPOD), and showed that 155 genes were differentially expressed by ≥2-fold in the pancreata of autoantibody-positive (AA+) at-risk individuals compared to healthy controls. Only 48 of these genes remained changed by ≥2-fold in the pancreata of established T1D patients. Pathway analysis of these genes showed a significant association with various immune pathways. We were able to validate the differential expression of eight disease-relevant genes by QPCR analysis: A significant upregulation of CADM2, and downregulation of TRPM5, CRH, PDK4, ANGPL4, CLEC4D, RSG16, and FCGR2B was confirmed in the pancreata of AA+ individuals versus controls. Studies have already implicated FCGR2B in the pathogenesis of disease in non-obese diabetic (NOD) mice. Here we showed that CADM2, TRPM5, PDK4, and ANGPL4 were similarly changed in the pancreata of pre-diabetic 12-week-old NOD mice compared to NOD.B10 controls, suggesting a possible role for these genes in the pathogenesis of both T1D and NOD disease. The loss of the leukocyte-specific gene, FCGR2B, in the pancreata of AA+ individuals, is particularly interesting, as it may serve as a potential whole blood biomarker of disease progression. To test this, we quantified FCGR2B expression in peripheral blood samples of T1D patients, and AA+ and AA- first-degree relatives of T1D patients enrolled in the TrialNet Pathway to Prevention study. We showed that FCGR2B was significantly reduced in the peripheral blood of AA+ individuals compared to AA- controls. Together, these findings demonstrate that gene expression analysis of pancreatic tissue and peripheral blood samples can be used to identify disease-relevant genes and pathways and potential biomarkers of disease progression in T1D.
Collapse
|
20
|
Noguchi GM, Huising MO. Integrating the inputs that shape pancreatic islet hormone release. Nat Metab 2019; 1:1189-1201. [PMID: 32694675 PMCID: PMC7378277 DOI: 10.1038/s42255-019-0148-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023]
Abstract
The pancreatic islet is a complex mini organ composed of a variety of endocrine cells and their support cells, which together tightly control blood glucose homeostasis. Changes in glucose concentration are commonly regarded as the chief signal controlling insulin-secreting beta cells, glucagon-secreting alpha cells and somatostatin-secreting delta cells. However, each of these cell types is highly responsive to a multitude of endocrine, paracrine, nutritional and neural inputs, which collectively shape the final endocrine output of the islet. Here, we review the principal inputs for each islet-cell type and the physiological circumstances in which these signals arise, through the prism of the insights generated by the transcriptomes of each of the major endocrine-cell types. A comprehensive integration of the factors that influence blood glucose homeostasis is essential to successfully improve therapeutic strategies for better diabetes management.
Collapse
Affiliation(s)
- Glyn M Noguchi
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, Davis, Davis, CA, USA
| | - Mark O Huising
- Department of Neurobiology, Physiology & Behavior, College of Biological Sciences, University of California, Davis, Davis, CA, USA.
- Department of Physiology & Membrane Biology, School of Medicine, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
21
|
Jiang Z, Rajamanickam S, Justice NJ. CRF signaling between neurons in the paraventricular nucleus of the hypothalamus (PVN) coordinates stress responses. Neurobiol Stress 2019; 11:100192. [PMID: 31516918 PMCID: PMC6732729 DOI: 10.1016/j.ynstr.2019.100192] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 08/07/2019] [Accepted: 08/08/2019] [Indexed: 12/22/2022] Open
Abstract
The importance of a precisely coordinated neuroendocrine, autonomic, and behavioral stress response was a primary theme at the Stress Neurobiology Workshop 2018, held in the beautiful setting of Banff Provincial Park in Alberta, Canada. Much of the research featured at this meeting reinforced the importance of appropriately responding to stress in order to avoid various neuropsychiatric pathologies, including Post-Traumatic Stress Disorder (PTSD), depression, and addiction. Corticotropin-Releasing Factor (CRF) neurons in the paraventricular nucleus of the hypothalamus (PVN) are central players in the stress response, integrating both external and visceral stress-relevant information, then directing neuroendocrine, autonomic and behavioral adaptations via endocrine and neural outputs of the PVN. The PVN contains a densely packed array of neuron types that respond to stress, including CRF neurons that activate the Hypothalamic-Pituitary-Adrenal (HPA) axis. Recently, identification of a new population of neurons in the PVN that express CRF Receptor 1 (CRFR1) has suggested that CRF release in the PVN signals to neighboring CRF responsive neurons, potentially functioning in HPA axis feedback, neuroendocrine coordination, and autonomic signaling. Here, we review our recent work characterizing an intra-PVN microcircuit in which locally released CRF release activates CRFR1+ neurons that make recurrent inhibitory GABAergic synapses onto CRF neurons to dampen excitability , therebylimiting HPA axis hyperactivity in response to stress and promoting stress recovery, which we presented in a poster session at the conference. We then discuss questions that have arisen following publication of our initial characterization of the microcircuit, regarding specific features of intra-PVN CRF signaling and its potential role in coordinating neuroendocrine, autonomic, and behavioral outputs of the PVN. Our presented work, as well as many of the presentations at the Stress Neurobiology Workshop 2018 together establish intra-PVN signaling as an important regulatory node in stress response pathways, which are central to the pathogenesis of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Zhiying Jiang
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Texas, 77030, USA
| | - Shivakumar Rajamanickam
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Texas, 77030, USA
| | - Nicholas J Justice
- The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Texas, 77030, USA
| |
Collapse
|
22
|
Prévost G, Picot M, Le Solliec MA, Arabo A, Berrahmoune H, El Mehdi M, Cherifi S, Benani A, Nédélec E, Gobet F, Brunel V, Leprince J, Lefebvre H, Anouar Y, Chartrel N. The neuropeptide 26RFa in the human gut and pancreas: potential involvement in glucose homeostasis. Endocr Connect 2019; 8:941-951. [PMID: 31234144 PMCID: PMC6612231 DOI: 10.1530/ec-19-0247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 06/12/2019] [Indexed: 01/25/2023]
Abstract
OBJECTIVE Recent studies performed in mice revealed that the neuropeptide 26RFa regulates glucose homeostasis by acting as an incretin and by increasing insulin sensitivity. However, in humans, an association between 26RFa and the regulation of glucose homeostasis is poorly documented. In this study, we have thus investigated in detail the distribution of 26RFa and its receptor, GPR103, in the gut and the pancreas, and determined the response of this peptidergic system to an oral glucose challenge in obese patients. DESIGN AND METHODS Distribution of 26RFa and GPR103 was examined by immunohistochemistry using gut and pancreas tissue sections. Circulating 26RFa was determined using a specific radioimmunoassay in plasma samples collected during an oral glucose tolerance test. RESULTS 26RFa and GPR103 are present all along the gut but are more abundant in the stomach and duodenum. In the stomach, the peptide and its receptor are highly expressed in the gastric glands, whereas in the duodenum, ileum and colon they are present in the enterocytes and the goblet cells. In the pancreatic islets, the 26RFa/GPR103 system is mostly present in the β cells. During an oral glucose tolerance test, plasma 26RFa profile is different between obese patients and healthy volunteers, and we found strong positive correlations between 26RFa blood levels and the BMI, and with various parameters of insulin secretion and insulin resistance. CONCLUSION The present data suggest an involvement of the 26RFa/GPR103 peptidergic system in the control of human glucose homeostasis.
Collapse
Affiliation(s)
- Gaëtan Prévost
- Normandie Univ, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Rouen, France
- Department of Endocrinology, Diabetes and Metabolic Diseases, Normandie Univ, UNIROUEN, Rouen University Hospital, Rouen, France
- Centre d’Investigation Clinique (CIC-CRB)-INSERM 1404, Rouen University Hospital, Rouen, France
- Correspondence should be addressed to G Prévost:
| | - Marie Picot
- Normandie Univ, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Rouen, France
| | - Marie-Anne Le Solliec
- Normandie Univ, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Rouen, France
| | - Arnaud Arabo
- Normandie Univ, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Rouen, France
| | - Hind Berrahmoune
- Normandie Univ, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Rouen, France
- Department of Endocrinology, Diabetes and Metabolic Diseases, Normandie Univ, UNIROUEN, Rouen University Hospital, Rouen, France
- Centre d’Investigation Clinique (CIC-CRB)-INSERM 1404, Rouen University Hospital, Rouen, France
| | - Mouna El Mehdi
- Normandie Univ, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Rouen, France
| | - Saloua Cherifi
- Normandie Univ, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Rouen, France
| | - Alexandre Benani
- Center for Taste and Feeding Behaviour, CNRS (UMR6265), INRA (UMR1324), Université de Bourgogne-Franche Comté, Dijon , France
| | - Emmanuelle Nédélec
- Center for Taste and Feeding Behaviour, CNRS (UMR6265), INRA (UMR1324), Université de Bourgogne-Franche Comté, Dijon , France
| | - Françoise Gobet
- Department of Anatomopathophysiology, Normandie Univ, UNIROUEN, Rouen University Hospital, Rouen, France
| | - Valéry Brunel
- Department of Biochemistry, Normandie Univ, UNIROUEN, Rouen University Hospital, Rouen, France
| | - Jérôme Leprince
- Normandie Univ, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Rouen, France
| | - Hervé Lefebvre
- Normandie Univ, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Rouen, France
- Department of Endocrinology, Diabetes and Metabolic Diseases, Normandie Univ, UNIROUEN, Rouen University Hospital, Rouen, France
- Centre d’Investigation Clinique (CIC-CRB)-INSERM 1404, Rouen University Hospital, Rouen, France
| | - Youssef Anouar
- Normandie Univ, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Rouen, France
| | - Nicolas Chartrel
- Normandie Univ, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N), Rouen, France
| |
Collapse
|
23
|
Prévost G, Arabo A, Le Solliec MA, Bons J, Picot M, Maucotel J, Berrahmoune H, El Mehdi M, Cherifi S, Benani A, Nédélec E, Coëffier M, Leprince J, Nordqvist A, Brunel V, Déchelotte P, Lefebvre H, Anouar Y, Chartrel N. Neuropeptide 26RFa (QRFP) is a key regulator of glucose homeostasis and its activity is markedly altered in obese/hyperglycemic mice. Am J Physiol Endocrinol Metab 2019; 317:E147-E157. [PMID: 31084498 DOI: 10.1152/ajpendo.00540.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recent studies have shown that the hypothalamic neuropeptide 26RFa regulates glucose homeostasis by acting as an incretin and increasing insulin sensitivity. In this study, we further characterized the role of the 26RFa/GPR103 peptidergic system in the global regulation of glucose homeostasis using a 26RFa receptor antagonist and also assessed whether a dysfunction of the 26RFa/GPR103 system occurs in obese hyperglycemic mice. First, we demonstrate that administration of the GPR103 antagonist reduces the global glucose-induced incretin effect and insulin sensitivity whereas, conversely, administration of exogenous 26RFa attenuates glucose-induced hyperglycemia. Using a mouse model of high-fat diet-induced obesity and hyperglycemia, we found a loss of the antihyperglcemic effect and insulinotropic activity of 26RFa, accompanied with a marked reduction of its insulin-sensitive effect. Interestingly, this resistance to 26RFa is associated with a downregulation of the 26RFa receptor in the pancreatic islets, and insulin target tissues. Finally, we observed that the production and release kinetics of 26RFa after an oral glucose challenge is profoundly altered in the high-fat mice. Altogether, the present findings support the view that 26RFa is a key regulator of glucose homeostasis whose activity is markedly altered under obese/hyperglycemic conditions.
Collapse
Affiliation(s)
- Gaëtan Prévost
- Normandie University, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N) , Rouen , France
- Normandie University, UNIROUEN, Rouen University Hospital, Department of Endocrinology, Diabetes and Metabolic Diseases, Rouen , France
| | - Arnaud Arabo
- Normandie University, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N) , Rouen , France
| | - Marie-Anne Le Solliec
- Normandie University, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N) , Rouen , France
| | - Justine Bons
- Normandie University, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N) , Rouen , France
- Normandie University, UNIROUEN, Rouen University Hospital, Department of Endocrinology, Diabetes and Metabolic Diseases, Rouen , France
| | - Marie Picot
- Normandie University, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N) , Rouen , France
| | - Julie Maucotel
- Normandie University, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N) , Rouen , France
| | - Hind Berrahmoune
- Normandie University, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N) , Rouen , France
- Normandie University, UNIROUEN, Rouen University Hospital, Department of Endocrinology, Diabetes and Metabolic Diseases, Rouen , France
| | - Mouna El Mehdi
- Normandie University, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N) , Rouen , France
| | - Saloua Cherifi
- Normandie University, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N) , Rouen , France
| | - Alexandre Benani
- Center for Taste and Feeding Behaviour, CNRS (UMR6265), INRA (UMR1324), Université de Bourgogne-Franche Comté , Dijon , France
| | - Emmanuelle Nédélec
- Center for Taste and Feeding Behaviour, CNRS (UMR6265), INRA (UMR1324), Université de Bourgogne-Franche Comté , Dijon , France
| | - Moïse Coëffier
- Normandie University, UNIROUEN, INSERM U1073 Nutrition, Inflammation and dysfunction of gut-brain axis, Rouen , France
- Normandie University, UNIROUEN, Rouen University Hospital, Department of Nutrition , Rouen , France
| | - Jérôme Leprince
- Normandie University, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N) , Rouen , France
| | - Anneli Nordqvist
- Cardiovascular Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Mölndal , Sweden
| | - Valéry Brunel
- Normandie University, UNIROUEN, Rouen University Hospital, Department of Biochemistry , Rouen , France
| | - Pierre Déchelotte
- Normandie University, UNIROUEN, INSERM U1073 Nutrition, Inflammation and dysfunction of gut-brain axis, Rouen , France
- Normandie University, UNIROUEN, Rouen University Hospital, Department of Nutrition , Rouen , France
| | - Hervé Lefebvre
- Normandie University, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N) , Rouen , France
- Normandie University, UNIROUEN, Rouen University Hospital, Department of Endocrinology, Diabetes and Metabolic Diseases, Rouen , France
| | - Youssef Anouar
- Normandie University, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N) , Rouen , France
| | - Nicolas Chartrel
- Normandie University, UNIROUEN, INSERM U1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication (DC2N) , Rouen , France
| |
Collapse
|
24
|
Inshaw JRJ, Cutler AJ, Burren OS, Stefana MI, Todd JA. Approaches and advances in the genetic causes of autoimmune disease and their implications. Nat Immunol 2018; 19:674-684. [PMID: 29925982 DOI: 10.1038/s41590-018-0129-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 04/04/2018] [Indexed: 12/18/2022]
Abstract
Genome-wide association studies are transformative in revealing the polygenetic basis of common diseases, with autoimmune diseases leading the charge. Although the field is just over 10 years old, advances in understanding the underlying mechanistic pathways of these conditions, which result from a dense multifactorial blend of genetic, developmental and environmental factors, have already been informative, including insights into therapeutic possibilities. Nevertheless, the challenge of identifying the actual causal genes and pathways and their biological effects on altering disease risk remains for many identified susceptibility regions. It is this fundamental knowledge that will underpin the revolution in patient stratification, the discovery of therapeutic targets and clinical trial design in the next 20 years. Here we outline recent advances in analytical and phenotyping approaches and the emergence of large cohorts with standardized gene-expression data and other phenotypic data that are fueling a bounty of discovery and improved understanding of human physiology.
Collapse
Affiliation(s)
- Jamie R J Inshaw
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Antony J Cutler
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Oliver S Burren
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - M Irina Stefana
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - John A Todd
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
| |
Collapse
|
25
|
Lim YJ, Kim JH, Pan JH, Kim JK, Park TS, Kim YJ, Lee JH, Kim JH. Naringin Protects Pancreatic β-Cells Against Oxidative Stress-Induced Apoptosis by Inhibiting Both Intrinsic and Extrinsic Pathways in Insulin-Deficient Diabetic Mice. Mol Nutr Food Res 2018; 62. [PMID: 29314619 DOI: 10.1002/mnfr.201700810] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/16/2017] [Indexed: 12/12/2022]
Abstract
SCOPE Oxidative stress has been suggested to play a central role in the pathogenesis of diabetes, as well as other metabolic disorders. Naringin, a major flavanone glycoside in citrus species, has been shown to display strong antioxidant potential in in vitro and in vivo models of oxidative stress; however, the underlying protective mechanisms in diabetes are unclear. METHODS AND RESULTS To study the protective effects and molecular mechanisms of naringin in preventing islet dysfunction and diabetes, we examined glucose homeostasis, β-cell apoptosis, and inflammatory response in insulin-deficient diabetic mice exposed to acute oxidative stress with streptozotocin (STZ). Naringin dose-dependently ameliorated hyperglycemia and islet dysfunction in insulin-deficient diabetic mice. Naringin counteracted STZ-induced β-cell apoptosis by inhibiting both the intrinsic (mitochondria-mediated) and extrinsic (death receptor-mediated) pathways. Furthermore, these protective effects were associated with suppression of DNA damage response and nuclear factor-kappa B- and mitogen-activated protein kinase-mediated signaling pathways, as well as reduction of reactive oxygen species accumulation and pro-inflammatory cytokine production in the pancreas. CONCLUSION Taken together, our study provides insights into the underlying mechanisms through which naringin protects the pancreatic β-cells against oxidative stress-induced apoptosis.
Collapse
Affiliation(s)
- Ye Jin Lim
- Department of Food and Biotechnology, Korea University, Sejong, South Korea
| | - Jung Ho Kim
- Department of Food and Biotechnology, Korea University, Sejong, South Korea
| | - Jeong Hoon Pan
- School of Human Environmental Sciences, University of Arkansas, Fayetteville, AR, USA
| | - Jae Kyeom Kim
- School of Human Environmental Sciences, University of Arkansas, Fayetteville, AR, USA
| | - Tae-Sik Park
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University of Medicine and Science, Inchon, South Korea
| | - Young Jun Kim
- Department of Food and Biotechnology, Korea University, Sejong, South Korea
| | - Jin Hyup Lee
- Department of Food and Biotechnology, Korea University, Sejong, South Korea
| | - Jun Ho Kim
- Department of Food and Biotechnology, Korea University, Sejong, South Korea
| |
Collapse
|
26
|
Tan LA, Vaughan JM, Perrin MH, Rivier JE, Sawchenko PE. Distribution of corticotropin-releasing factor (CRF) receptor binding in the mouse brain using a new, high-affinity radioligand, [125I]-PD-Sauvagine. J Comp Neurol 2017; 525:3840-3864. [DOI: 10.1002/cne.24307] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/15/2017] [Accepted: 08/16/2017] [Indexed: 01/30/2023]
Affiliation(s)
- Laura A. Tan
- Laboratory of Neuronal Structure and Function; The Salk Institute for Biological Studies; La Jolla CA
| | - Joan M. Vaughan
- Laboratory of Neuronal Structure and Function; The Salk Institute for Biological Studies; La Jolla CA
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies; La Jolla CA
| | - Marilyn H. Perrin
- Laboratory of Neuronal Structure and Function; The Salk Institute for Biological Studies; La Jolla CA
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies; La Jolla CA
| | - Jean E. Rivier
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies; La Jolla CA
| | - Paul E. Sawchenko
- Laboratory of Neuronal Structure and Function; The Salk Institute for Biological Studies; La Jolla CA
| |
Collapse
|
27
|
van der Meulen T, Mawla AM, DiGruccio MR, Adams MW, Nies V, Dólleman S, Liu S, Ackermann AM, Cáceres E, Hunter AE, Kaestner KH, Donaldson CJ, Huising MO. Virgin Beta Cells Persist throughout Life at a Neogenic Niche within Pancreatic Islets. Cell Metab 2017; 25:911-926.e6. [PMID: 28380380 PMCID: PMC8586897 DOI: 10.1016/j.cmet.2017.03.017] [Citation(s) in RCA: 162] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 02/14/2017] [Accepted: 03/21/2017] [Indexed: 12/21/2022]
Abstract
Postnatal maintenance or regeneration of pancreatic beta cells is considered to occur exclusively via the replication of existing beta cells, but clinically meaningful restoration of human beta cell mass by proliferation has never been achieved. We discovered a population of immature beta cells that is present throughout life and forms from non-beta precursors at a specialized micro-environment or "neogenic niche" at the islet periphery. These cells express insulin, but lack other key beta cell markers, and are transcriptionally immature, incapable of sensing glucose, and unable to support calcium influx. They constitute an intermediate stage in the transdifferentiation of alpha cells to cells that are functionally indistinguishable from conventional beta cells. We thus identified a lifelong source of new beta cells at a specialized site within healthy islets. By comparing co-existing immature and mature beta cells within healthy islets, we stand to learn how to mature insulin-expressing cells into functional beta cells.
Collapse
Affiliation(s)
- Talitha van der Meulen
- Department of Neurobiology, Physiology, and Behavior, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA
| | - Alex M Mawla
- Department of Neurobiology, Physiology, and Behavior, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA
| | - Michael R DiGruccio
- Department of Neurobiology, Physiology, and Behavior, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA
| | - Michael W Adams
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Vera Nies
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Sophie Dólleman
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Siming Liu
- Department of Neurobiology, Physiology, and Behavior, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA
| | - Amanda M Ackermann
- Division of Endocrinology and Diabetes, The Children's Hospital of Philadelphia, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Elena Cáceres
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Anna E Hunter
- Department of Neurobiology, Physiology, and Behavior, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cynthia J Donaldson
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Mark O Huising
- Department of Neurobiology, Physiology, and Behavior, College of Biological Sciences, University of California, Davis, Davis, CA 95616, USA; Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
28
|
Yu C, Li M, Wang Y, Liu Y, Yan C, Pan J, Liu J, Cui S. MiR-375 Mediates CRH Signaling Pathway in Inhibiting E2 Synthesis in Porcine Ovary. Reproduction 2016; 153:REP-16-0323. [PMID: 27756810 DOI: 10.1530/rep-16-0323] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/17/2016] [Indexed: 12/31/2022]
Abstract
The corticotropin-releasing hormone (CRH) signaling system is involved in numbers of stress-related physiological and pathological responses,including its inhibiting effects on estradiol (E2) synthesis and follicular development in the ovary. In addition, there are reports that microRNAs (miRNAs) can control the function of animal reproductive system. The aim of present study was to investigate the functions of miR-375 and the relationship between miR-375 and CRH signaling molecules in the porcine ovary. First, our common PCR results show that miR-375 and the CRH receptor 1 (CRHR1) are expressed in porcine ovary, whereas CRH receptor 2 (CRHR2) is not detected. We further have located the cell types of miR-375 and CRHR1 by in situ hybridization (ISH), and the results show that miR-375 is located only in the granulosa cells, whereas CRHR1 is positive in all of granulosa cells and oocytes, inferring that miR-375 and CRHR1 are co-localized in granulosa cells. Second, we show that overexpression of miR-375 in cultured granulosa cells suppresses the E2 production, while miR-375 knockdown demonstrates the opposite result. Besides, our in vitro results demonstrate that miR-375 mediates the signaling pathway of CRH inhibiting E2 synthesis. Finally, our data show that the action of miR-375 is accomplished by directly binding to the 3'UTR of specificity protein1 (SP1) mRNA to decrease the SP1 protein level. Thus, we conclude that miR-375 is a key factor in regulating E2 synthesis by mediating the CRH signaling pathway.
Collapse
Affiliation(s)
- Chulin Yu
- C Yu, Department of Animal Physiology, College of Biological Sciences,China Agricultural University, Beijing, China
| | - Meiling Li
- M Li, Department of Animal Physiology, College of Biological Sciences,China Agricultural University, Beijing, China
| | - Yue Wang
- Y Wang, Department of Animal Physiology, College of Biological Sciences,China Agricultural University, Beijing, China
| | - Ying Liu
- Y Liu, Department of Animal Physiology, College of Biological Sciences,China Agricultural University, Beijing, China
| | - Chengzhi Yan
- C Yan, Department of Animal Physiology, College of Biological Sciences,China Agricultural University, Beijing, China
| | - Jirong Pan
- J Pan, Department of Animal Physiology, College of Biological Sciences,China Agricultural University, Beijing, China
| | - Jiali Liu
- J Liu, Department of Animal Physiology, College of Biological Sciences,China Agricultural University, Beijing, China
| | - Sheng Cui
- S Cui, Department of Animal Physiology, College of Biological Sciences,China Agricultural University, Beijing, China
| |
Collapse
|
29
|
Comprehensive alpha, beta and delta cell transcriptomes reveal that ghrelin selectively activates delta cells and promotes somatostatin release from pancreatic islets. Mol Metab 2016; 5:449-458. [PMID: 27408771 PMCID: PMC4921781 DOI: 10.1016/j.molmet.2016.04.007] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/14/2016] [Accepted: 04/21/2016] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Complex local crosstalk amongst endocrine cells within the islet ensures tight coordination of their endocrine output. This is illustrated by the recent demonstration that the negative feedback control by delta cells within pancreatic islets determines the homeostatic set-point for plasma glucose during mouse postnatal development. However, the close association of islet endocrine cells that facilitates paracrine crosstalk also complicates the distinction between effects mediated directly on beta cells from indirect effects mediated via local intermediates, such as somatostatin from delta cells. METHODS To resolve this problem, we generated reporter mice that allow collection of pure pancreatic delta cells along with alpha and beta cells from the same islets and generated comprehensive transcriptomes for each islet endocrine cell type. These transcriptomes afford an unparalleled view of the receptors expressed by delta, alpha and beta cells, and allow the prediction of which signal targets which endocrine cell type with great accuracy. RESULTS From these transcriptomes, we discovered that the ghrelin receptor is expressed exclusively by delta cells within the islet, which was confirmed by fluorescent in situ hybridization and qPCR. Indeed, ghrelin increases intracellular calcium in delta cells in intact mouse islets, measured by GCaMP6 and robustly potentiates glucose-stimulated somatostatin secretion on mouse and human islets in both static and perfusion assays. In contrast, des-acyl-ghrelin at the same dose had no effect on somatostatin secretion and did not block the actions of ghrelin. CONCLUSIONS These results offer a straightforward explanation for the well-known insulinostatic actions of ghrelin. Rather than engaging beta cells directly, ghrelin engages delta cells to promote local inhibitory feedback that attenuates insulin release. These findings illustrate the power of our approach to resolve some of the long-standing conundrums with regard to the rich feedback that occurs within the islet that is integral to islet physiology and therefore highly relevant to diabetes.
Collapse
|
30
|
Barkai U, Rotem A, de Vos P. Survival of encapsulated islets: More than a membrane story. World J Transplant 2016; 6:69-90. [PMID: 27011906 PMCID: PMC4801806 DOI: 10.5500/wjt.v6.i1.69] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/02/2015] [Accepted: 12/20/2015] [Indexed: 02/05/2023] Open
Abstract
At present, proven clinical treatments but no cures are available for diabetes, a global epidemic with a huge economic burden. Transplantation of islets of Langerhans by their infusion into vascularized organs is an experimental clinical protocol, the first approach to attain cure. However, it is associated with lifelong use of immunosuppressants. To overcome the need for immunosuppression, islets are encapsulated and separated from the host immune system by a permselective membrane. The lead material for this application is alginate which was tested in many animal models and a few clinical trials. This review discusses all aspects related to the function of transplanted encapsulated islets such as the basic requirements from a permselective membrane (e.g., allowable hydrodynamic radii, implications of the thickness of the membrane and relative electrical charge). Another aspect involves adequate oxygen supply, which is essential for survival/performance of transplanted islets, especially when using large retrievable macro-capsules implanted in poorly oxygenated sites like the subcutis. Notably, islets can survive under low oxygen tension and are physiologically active at > 40 Torr. Surprisingly, when densely crowded, islets are fully functional under hyperoxic pressure of up to 500 Torr (> 300% of atmospheric oxygen tension). The review also addresses an additional category of requirements for optimal performance of transplanted islets, named auxiliary technologies. These include control of inflammation, apoptosis, angiogenesis, and the intra-capsular environment. The review highlights that curing diabetes with a functional bio-artificial pancreas requires optimizing all of these aspects, and that significant advances have already been made in many of them.
Collapse
|
31
|
GUO H, FANG Q, HUO Y, ZHANG Y, ZHANG J. Social dominance-related major urinary proteins and the regulatory mechanism in mice. Integr Zool 2015; 10:543-54. [DOI: 10.1111/1749-4877.12165] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Huifen GUO
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture; Institute of Zoology, Chinese Academy of Sciences; Beijing China
| | - Qi FANG
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture; Institute of Zoology, Chinese Academy of Sciences; Beijing China
| | - Ying HUO
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture; Institute of Zoology, Chinese Academy of Sciences; Beijing China
| | - Yaohua ZHANG
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture; Institute of Zoology, Chinese Academy of Sciences; Beijing China
| | - Jianxu ZHANG
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture; Institute of Zoology, Chinese Academy of Sciences; Beijing China
| |
Collapse
|
32
|
Regulation of Glucose Homeostasis by Glucocorticoids. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015. [PMID: 26215992 DOI: 10.1007/978-1-4939-2895-8_5] [Citation(s) in RCA: 402] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glucocorticoids are steroid hormones that regulate multiple aspects of glucose homeostasis. Glucocorticoids promote gluconeogenesis in liver, whereas in skeletal muscle and white adipose tissue they decrease glucose uptake and utilization by antagonizing insulin response. Therefore, excess glucocorticoid exposure causes hyperglycemia and insulin resistance. Glucocorticoids also regulate glycogen metabolism. In liver, glucocorticoids increase glycogen storage, whereas in skeletal muscle they play a permissive role for catecholamine-induced glycogenolysis and/or inhibit insulin-stimulated glycogen synthesis. Moreover, glucocorticoids modulate the function of pancreatic α and β cells to regulate the secretion of glucagon and insulin, two hormones that play a pivotal role in the regulation of blood glucose levels. Overall, the major glucocorticoid effect on glucose homeostasis is to preserve plasma glucose for brain during stress, as transiently raising blood glucose is important to promote maximal brain function. In this chapter we will discuss the current understanding of the mechanisms underlying different aspects of glucocorticoid-regulated mammalian glucose homeostasis.
Collapse
|
33
|
Prévost G, Jeandel L, Arabo A, Coëffier M, El Ouahli M, Picot M, Alexandre D, Gobet F, Leprince J, Berrahmoune H, Déchelotte P, Malagon M, Bonner C, Kerr-Conte J, Chigr F, Lefebvre H, Anouar Y, Chartrel N. Hypothalamic Neuropeptide 26RFa Acts as an Incretin to Regulate Glucose Homeostasis. Diabetes 2015; 64:2805-16. [PMID: 25858563 DOI: 10.2337/db14-1864] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 04/02/2015] [Indexed: 11/13/2022]
Abstract
26RFa is a hypothalamic neuropeptide that promotes food intake. 26RFa is upregulated in obese animal models, and its orexigenic activity is accentuated in rodents fed a high-fat diet, suggesting that this neuropeptide might play a role in the development and maintenance of the obese status. As obesity is frequently associated with type 2 diabetes, we investigated whether 26RFa may be involved in the regulation of glucose homeostasis. In the current study, we show a moderate positive correlation between plasma 26RFa levels and plasma insulin in patients with diabetes. Plasma 26RFa concentration also increases in response to an oral glucose tolerance test. In addition, we found that 26RFa and its receptor GPR103 are present in human pancreatic β-cells as well as in the gut. In mice, 26RFa attenuates the hyperglycemia induced by a glucose load, potentiates insulin sensitivity, and increases plasma insulin concentrations. Consistent with these data, 26RFa stimulates insulin production by MIN6 insulinoma cells. Finally, we show, using in vivo and in vitro approaches, that a glucose load induces a massive secretion of 26RFa by the small intestine. Altogether, the present data indicate that 26RFa acts as an incretin to regulate glucose homeostasis.
Collapse
Affiliation(s)
- Gaëtan Prévost
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, Rouen, France Department of Endocrinology, Diabetes and Metabolic Diseases, Institute for Research and Innovation in Biomedicine, University Hospital of Rouen, Rouen, France Normandy University, Caen, France University of Rouen, Rouen, France
| | - Lydie Jeandel
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, Rouen, France Normandy University, Caen, France University of Rouen, Rouen, France
| | - Arnaud Arabo
- Normandy University, Caen, France University of Rouen, Rouen, France
| | - Moïse Coëffier
- Normandy University, Caen, France University of Rouen, Rouen, France INSERM U1073, Institute for Research and Innovation in Biomedicine, Rouen, France Department of Nutrition, University Hospital of Rouen, Rouen, France
| | - Mariama El Ouahli
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, Rouen, France Normandy University, Caen, France University of Rouen, Rouen, France Biological Engineering Laboratory, Life Sciences, Sultan Moulay Slimane University, Beni-Mellal, Morocco
| | - Marie Picot
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, Rouen, France Normandy University, Caen, France University of Rouen, Rouen, France
| | - David Alexandre
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, Rouen, France Normandy University, Caen, France University of Rouen, Rouen, France
| | - Françoise Gobet
- Normandy University, Caen, France University of Rouen, Rouen, France Department of Pathology, University Hospital of Rouen, Rouen, France
| | - Jérôme Leprince
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, Rouen, France Normandy University, Caen, France University of Rouen, Rouen, France
| | - Hind Berrahmoune
- Department of Endocrinology, Diabetes and Metabolic Diseases, Institute for Research and Innovation in Biomedicine, University Hospital of Rouen, Rouen, France Normandy University, Caen, France University of Rouen, Rouen, France
| | - Pierre Déchelotte
- Normandy University, Caen, France University of Rouen, Rouen, France INSERM U1073, Institute for Research and Innovation in Biomedicine, Rouen, France Department of Nutrition, University Hospital of Rouen, Rouen, France
| | - Maria Malagon
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba/Reina Sofía University Hospital, University of Cordoba, Cordoba, Spain
| | - Caroline Bonner
- INSERM U859, Biotherapies of Diabetes, Faculty of Medicine, University of Lille, Lille, France
| | - Julie Kerr-Conte
- INSERM U859, Biotherapies of Diabetes, Faculty of Medicine, University of Lille, Lille, France
| | - Fatiha Chigr
- Biological Engineering Laboratory, Life Sciences, Sultan Moulay Slimane University, Beni-Mellal, Morocco
| | - Hervé Lefebvre
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, Rouen, France Department of Endocrinology, Diabetes and Metabolic Diseases, Institute for Research and Innovation in Biomedicine, University Hospital of Rouen, Rouen, France Normandy University, Caen, France University of Rouen, Rouen, France
| | - Youssef Anouar
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, Rouen, France Normandy University, Caen, France University of Rouen, Rouen, France
| | - Nicolas Chartrel
- INSERM U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, Rouen, France Normandy University, Caen, France University of Rouen, Rouen, France
| |
Collapse
|
34
|
Urocortin3 mediates somatostatin-dependent negative feedback control of insulin secretion. Nat Med 2015; 21:769-76. [PMID: 26076035 PMCID: PMC4496282 DOI: 10.1038/nm.3872] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 04/30/2015] [Indexed: 12/15/2022]
Abstract
The peptide hormone urocortin3 (Ucn3) is abundantly expressed by mature beta cells, yet its physiological role is unknown. Here we demonstrate that Ucn3 is stored and co-released with insulin and potentiates glucose-stimulated somatostatin secretion via cognate receptors on delta cells. Further, we found that islets lacking endogenous Ucn3 have fewer delta cells, reduced somatostatin content, impaired somatostatin secretion, and exaggerated insulin release, and that these defects are rectified by treatment with synthetic Ucn3 in vitro. Our observations indicate that the paracrine actions of Ucn3 activate a negative feedback loop that promotes somatostatin release to ensure the timely reduction of insulin secretion upon normalization of plasma glucose. Moreover, Ucn3 is markedly depleted from beta cells in mouse and macaque models of diabetes and in human diabetic islets. This suggests that Ucn3 is a key contributor to stable glycemic control, whose reduction during diabetes aggravates glycemic volatility and contributes to the pathophysiology of this disease.
Collapse
|
35
|
Hao K, Kong FP, Gao YQ, Tang JW, Chen J, Evans AM, Lightman SL, Chen XQ, Du JZ. Inactivation of corticotropin-releasing hormone-induced insulinotropic role by high-altitude hypoxia. Diabetes 2015; 64:785-95. [PMID: 25277397 DOI: 10.2337/db14-0500] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We have shown that hypoxia reduces plasma insulin, which correlates with corticotropin-releasing hormone (CRH) receptor 1 (CRHR1) in rats, but the mechanism remains unclear. Here, we report that hypobaric hypoxia at an altitude of 5,000 m for 8 h enhances rat plasma CRH, corticosterone, and glucose levels, whereas the plasma insulin and pancreatic ATP/ADP ratio is reduced. In islets cultured under normoxia, CRH stimulated insulin release in a glucose- and CRH-level-dependent manner by activating CRHR1 and thus the cAMP-dependent protein kinase pathway and calcium influx through L-type channels. In islets cultured under hypoxia, however, the insulinotropic effect of CRH was inactivated due to reduced ATP and cAMP and coincident loss of intracellular calcium oscillations. Serum and glucocorticoid-inducible kinase 1 (SGK1) also played an inhibitory role. In human volunteers rapidly ascended to 3,860 m, plasma CRH and glucose levels increased without a detectable change in plasma insulin. By contrast, volunteers with acute mountain sickness (AMS) exhibited a marked decrease in HOMA insulin sensitivity (HOMA-IS) and enhanced plasma CRH. In conclusion, hypoxia may attenuate the CRH-insulinotropic effect by reducing cellular ATP/ADP ratio, cAMP and calcium influx, and upregulated SGK1. Hypoxia may not affect HOMA-IS in healthy volunteers but reduces it in AMS volunteers.
Collapse
Affiliation(s)
- Ke Hao
- Division of Neurobiology and Physiology, Department of Basic Medical Sciences, School of Medicine, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Fan-Ping Kong
- Division of Neurobiology and Physiology, Department of Basic Medical Sciences, School of Medicine, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Yu-Qi Gao
- Department of Pathophysiology and High Altitude Physiology, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China
| | - Jia-Wei Tang
- Division of Neurobiology and Physiology, Department of Basic Medical Sciences, School of Medicine, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Jian Chen
- Department of Pathophysiology and High Altitude Physiology, College of High Altitude Military Medicine, Third Military Medical University, Chongqing, China
| | - A Mark Evans
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, U.K
| | - Stafford L Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, U.K
| | - Xue-Qun Chen
- Division of Neurobiology and Physiology, Department of Basic Medical Sciences, School of Medicine, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| | - Ji-Zeng Du
- Division of Neurobiology and Physiology, Department of Basic Medical Sciences, School of Medicine, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang University, Hangzhou, China
| |
Collapse
|
36
|
Prothiwa M, Syed I, Huising MO, van der Meulen T, Donaldson CJ, Trauger SA, Kahn BB, Saghatelian A. Data-driven synthesis of proteolysis-resistant peptide hormones. J Am Chem Soc 2014; 136:17710-3. [PMID: 25496053 DOI: 10.1021/ja5065735] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Peptide hormones are key physiological regulators, and many would make terrific drugs; however, the therapeutic use of peptides is limited by poor metabolism including rapid proteolysis. To develop novel proteolysis-resistant peptide hormone analogs, we utilize a strategy that relies on data from simple mass spectrometry experiments to guide the chemical synthesis of proteolysis-resistant analogs (i.e., data-driven synthesis). Application of this strategy to oxyntomodulin (OXM), a peptide hormone that stimulates insulin secretion from islets and lowers blood glucose in vivo, defined the OXM cleavage site in serum, and this information was used to synthesize a proteolysis-resistant OXM analog (prOXM). prOXM and OXM have similar activity in binding and glucose stimulated-insulin secretion assays. Furthermore, prOXM is also active in vivo. prOXM reduces basal glucose levels and improves glucose tolerance in mice. The discovery of prOXM suggests that proteolysis-resistant variants of other important peptide hormones can also be found using this strategy to increase the number of candidate therapeutic peptides.
Collapse
Affiliation(s)
- Michaela Prothiwa
- Department of Chemistry and Chemical Biology, Harvard University , Cambridge, Massachusetts 02138, United States
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Blaabjerg L, Christensen GL, Matsumoto M, van der Meulen T, Huising MO, Billestrup N, Vale WW. CRFR1 activation protects against cytokine-induced β-cell death. J Mol Endocrinol 2014; 53:417-27. [PMID: 25324488 PMCID: PMC4518718 DOI: 10.1530/jme-14-0056] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
During the development of diabetes β-cells are exposed to elevated concentrations of proinflammatory cytokines, TNFα and IL1β, which in vitro induce β-cell death. The class B G-protein-coupled receptors (GPCRs): corticotropin-releasing factor receptor 1 (CRFR1) and CRFR2 are expressed in pancreatic islets. As downstream signaling by other class B GPCRs can protect against cytokine-induced β-cell apoptosis, we evaluated the protective potential of CRFR activation in β-cells in a pro-inflammatory setting. CRFR1/CRFR2 ligands activated AKT and CRFR1 signaling and reduced apoptosis in human islets. In rat and mouse insulin-secreting cell lines (INS-1 and MIN6), CRFR1 agonists upregulated insulin receptor substrate 2 (IRS2) expression, increased AKT activation, counteracted the cytokine-mediated decrease in BAD phosphorylation, and inhibited apoptosis. The anti-apoptotic signaling was dependent on prolonged exposure to corticotropin-releasing factor family peptides and followed PKA-mediated IRS2 upregulation. This indicates that CRFR signaling counteracts proinflammatory cytokine-mediated apoptotic pathways through upregulation of survival signaling in β-cells. Interestingly, CRFR signaling also counteracted basal apoptosis in both cultured INS-1 cells and intact human islets.
Collapse
Affiliation(s)
- Lykke Blaabjerg
- Clayton Foundation Laboratories for Peptide BiologySalk Institute, 10100 North Torrey Pines Road, La Jolla, California 92037, USACellular and Metabolic Research SectionDepartment of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark Clayton Foundation Laboratories for Peptide BiologySalk Institute, 10100 North Torrey Pines Road, La Jolla, California 92037, USACellular and Metabolic Research SectionDepartment of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Gitte L Christensen
- Clayton Foundation Laboratories for Peptide BiologySalk Institute, 10100 North Torrey Pines Road, La Jolla, California 92037, USACellular and Metabolic Research SectionDepartment of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Masahito Matsumoto
- Clayton Foundation Laboratories for Peptide BiologySalk Institute, 10100 North Torrey Pines Road, La Jolla, California 92037, USACellular and Metabolic Research SectionDepartment of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Talitha van der Meulen
- Clayton Foundation Laboratories for Peptide BiologySalk Institute, 10100 North Torrey Pines Road, La Jolla, California 92037, USACellular and Metabolic Research SectionDepartment of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Mark O Huising
- Clayton Foundation Laboratories for Peptide BiologySalk Institute, 10100 North Torrey Pines Road, La Jolla, California 92037, USACellular and Metabolic Research SectionDepartment of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Nils Billestrup
- Clayton Foundation Laboratories for Peptide BiologySalk Institute, 10100 North Torrey Pines Road, La Jolla, California 92037, USACellular and Metabolic Research SectionDepartment of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Wylie W Vale
- Clayton Foundation Laboratories for Peptide BiologySalk Institute, 10100 North Torrey Pines Road, La Jolla, California 92037, USACellular and Metabolic Research SectionDepartment of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| |
Collapse
|
38
|
Riera CE, Huising MO, Follett P, Leblanc M, Halloran J, Van Andel R, de Magalhaes Filho CD, Merkwirth C, Dillin A. TRPV1 pain receptors regulate longevity and metabolism by neuropeptide signaling. Cell 2014; 157:1023-36. [PMID: 24855942 DOI: 10.1016/j.cell.2014.03.051] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 01/06/2014] [Accepted: 03/20/2014] [Indexed: 01/11/2023]
Abstract
The sensation of pain is associated with increased mortality, but it is unknown whether pain perception can directly affect aging. We find that mice lacking TRPV1 pain receptors are long-lived, displaying a youthful metabolic profile at old age. Loss of TRPV1 inactivates a calcium-signaling cascade that ends in the nuclear exclusion of the CREB-regulated transcriptional coactivator CRTC1 within pain sensory neurons originating from the spinal cord. In long-lived TRPV1 knockout mice, CRTC1 nuclear exclusion decreases production of the neuropeptide CGRP from sensory endings innervating the pancreatic islets, subsequently promoting insulin secretion and metabolic health. In contrast, CGRP homeostasis is disrupted with age in wild-type mice, resulting in metabolic decline. We show that pharmacologic inactivation of CGRP receptors in old wild-type animals can restore metabolic health. These data suggest that ablation of select pain sensory receptors or the inhibition of CGRP are associated with increased metabolic health and control longevity.
Collapse
Affiliation(s)
- Céline E Riera
- Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; The Glenn Center for Aging Research, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Mark O Huising
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Patricia Follett
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; The Glenn Center for Aging Research, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Mathias Leblanc
- The Glenn Center for Aging Research, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jonathan Halloran
- Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Roger Van Andel
- Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Carsten Merkwirth
- The Glenn Center for Aging Research, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Andrew Dillin
- Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; The Glenn Center for Aging Research, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
39
|
Benner C, van der Meulen T, Cacéres E, Tigyi K, Donaldson CJ, Huising MO. The transcriptional landscape of mouse beta cells compared to human beta cells reveals notable species differences in long non-coding RNA and protein-coding gene expression. BMC Genomics 2014; 15:620. [PMID: 25051960 PMCID: PMC4124169 DOI: 10.1186/1471-2164-15-620] [Citation(s) in RCA: 209] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 07/10/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Insulin producing beta cell and glucagon producing alpha cells are colocalized in pancreatic islets in an arrangement that facilitates the coordinated release of the two principal hormones that regulate glucose homeostasis and prevent both hypoglycemia and diabetes. However, this intricate organization has also complicated the determination of the cellular source(s) of the expression of genes that are detected in the islet. This reflects a significant gap in our understanding of mouse islet physiology, which reduces the effectiveness by which mice model human islet disease. RESULTS To overcome this challenge, we generated a bitransgenic reporter mouse that faithfully labels all beta and alpha cells in mouse islets to enable FACS-based purification and the generation of comprehensive transcriptomes of both populations. This facilitates systematic comparison across thousands of genes between the two major endocrine cell types of the islets of Langerhans whose principal hormones are of cardinal importance for glucose homeostasis. Our data leveraged against similar data for human beta cells reveal a core common beta cell transcriptome of 9900+ genes. Against the backdrop of overall similar beta cell transcriptomes, we describe marked differences in the repertoire of receptors and long non-coding RNAs between mouse and human beta cells. CONCLUSIONS The comprehensive mouse alpha and beta cell transcriptomes complemented by the comparison of the global (dis)similarities between mouse and human beta cells represent invaluable resources to boost the accuracy by which rodent models offer guidance in finding cures for human diabetes.
Collapse
Affiliation(s)
- Christopher Benner
- />Razzavi Newman Integrated Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Talitha van der Meulen
- />Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Elena Cacéres
- />Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Kristof Tigyi
- />Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Cynthia J Donaldson
- />Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
| | - Mark O Huising
- />Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037 USA
- />Department of Neurobiology, Physiology & Behavior, University of California, One Shields Avenue, 180 Briggs Hall, Davis, CA 95616 USA
| |
Collapse
|
40
|
Gragnoli C. Hypothesis of the neuroendocrine cortisol pathway gene role in the comorbidity of depression, type 2 diabetes, and metabolic syndrome. APPLICATION OF CLINICAL GENETICS 2014; 7:43-53. [PMID: 24817815 PMCID: PMC4012344 DOI: 10.2147/tacg.s39993] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Depression, type 2 diabetes (T2D), and metabolic syndrome (MetS) are often comorbid. Depression per se increases the risk for T2D by 60%. This risk is not accounted for by the use of antidepressant therapy. Stress causes hyperactivation of the hypothalamic–pituitary–adrenal (HPA) axis, by triggering the hypothalamic corticotropin-releasing hormone (CRH) secretion, which stimulates the anterior pituitary to release the adrenocorticotropin hormone (ACTH), which causes the adrenal secretion of cortisol. Depression is associated with an increased level of cortisol, and CRH and ACTH at inappropriately “normal” levels, that is too high compared to their expected lower levels due to cortisol negative feedback. T2D and MetS are also associated with hypercortisolism. High levels of cortisol can impair mood as well as cause hyperglycemia and insulin resistance and other traits typical of T2D and MetS. We hypothesize that HPA axis hyperactivation may be due to variants in the genes of the CRH receptors (CRHR1, CRHR2), corticotropin receptors (or melanocortin receptors, MC1R-MC5R), glucocorticoid receptor (NR3C1), mineralocorticoid receptor (NR3C2), and of the FK506 binding protein 51 (FKBP5), and that these variants may be partially responsible for the clinical association of depression, T2D and MetS. In this review, we will focus on the correlation of stress, HPA axis hyperactivation, and the possible genetic role of the CRHR1, CRHR2, MCR1–5, NR3C1, and NR3C2 receptors and FKBP5 in the susceptibility to the comorbidity of depression, T2D, and MetS. New studies are needed to confirm the hypothesized role of these genes in the clinical association of depression, T2D, and MetS.
Collapse
Affiliation(s)
- Claudia Gragnoli
- Center for Biotechnology and Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA ; Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, Rome, Italy
| |
Collapse
|
41
|
Ludwig B, Barthel A, Reichel A, Block NL, Ludwig S, Schally AV, Bornstein SR. Modulation of the pancreatic islet-stress axis as a novel potential therapeutic target in diabetes mellitus. VITAMINS AND HORMONES 2014; 95:195-222. [PMID: 24559919 DOI: 10.1016/b978-0-12-800174-5.00008-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Loss of pancreatic islet function and insulin-producing beta cell mass is a central hallmark in the pathogenesis of both type 1 and type 2 diabetes. While in type 1 diabetes this phenomenon is due to an extensive destruction of beta cells caused by an autoimmune process, the mechanisms resulting in beta cell failure in type 2 diabetes are different and less clear. Also, beta cell destruction in type 1 diabetes occurs early and is the initial step in the pathogenetic process, while beta cell loss in type 2 diabetes after an initial phase of hyperinsulinemia due to the underlying insulin resistance occurs relatively late and it is less pronounced. Since diabetes mellitus is the most frequent endocrine disease, with an increasing high prevalence worldwide, huge efforts have been made over the past many decades to identify predisposing genetic, environmental, and nutritional factors in order to develop effective strategies to prevent the disease. In parallel, extensive studies in different cell systems and animal models have helped to elucidate our understanding of the physiologic function of islets and to gain insight into the immunological and non-immunological mechanisms of beta cell destruction and failure. Furthermore, currently emerging concepts of beta cell regeneration (e.g., the restoration of the beta cell pool by regenerative, proliferative and antiapoptotic processes, and recovery of physiologic islet function) apparently is yielding the first promising results. Recent insights into the complex endocrine and paracrine mechanisms regulating the physiologic function of pancreatic islets, as well as beta cell life and death, constitute an essential part of this new and exciting area of diabetology. For example, understanding of the physiological role of glucagon-like peptide 1 has resulted in the successful clinical implementation of incretin-based therapies over the last years. Further, recent data suggesting paracrine effects of growth hormone-releasing hormone and corticotropin-releasing hormone on the regulation of pancreatic islet function, survival, and proliferation as well as on local glucocorticoid metabolism provide evidence for a potential role of the pancreatic islet-stress axis in the pathophysiology of diabetes mellitus. In this chapter, we provide a comprehensive overview of current preventive and regenerative concepts as a basis for the development of novel therapeutic approaches to the treatment of diabetes mellitus. A particular focus is given on the potential of the pancreatic islet-stress axis in the development of novel regenerative strategies.
Collapse
Affiliation(s)
- Barbara Ludwig
- Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany; The Paul Langerhans Institute, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany.
| | - Andreas Barthel
- Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany; Endokrinologikum Ruhr, Bochum, Germany
| | - Andreas Reichel
- Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Norman L Block
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Medicine, Division of Hematology-Oncology, University of Miami Miller School of Medicine, Miami, Florida, USA; Veterans Administration Medical Center, Miami, Florida, USA
| | - Stefan Ludwig
- Department of Visceral, Thorax and Vascular Surgery, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Andrew V Schally
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Medicine, Division of Endocrinology, University of Miami Miller School of Medicine, Miami, Florida, USA; Department of Medicine, Division of Hematology-Oncology, University of Miami Miller School of Medicine, Miami, Florida, USA; Veterans Administration Medical Center, Miami, Florida, USA
| | - Stefan R Bornstein
- Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany; The Paul Langerhans Institute, Dresden, Germany; Center for Regenerative Therapies Dresden, Dresden University of Technology, Dresden, Germany
| |
Collapse
|
42
|
Sakamoto R, Matsubara E, Nomura M, Wang L, Kawahara Y, Yanase T, Nawata H, Takayanagi R. Roles for corticotropin-releasing factor receptor type 1 in energy homeostasis in mice. Metabolism 2013; 62:1739-48. [PMID: 24054833 DOI: 10.1016/j.metabol.2013.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 08/09/2013] [Accepted: 08/13/2013] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Expression of corticotropin-releasing factor type 1 receptor (CRFR1) has been shown on pancreatic β cells, and its activation potentiates glucose-stimulated insulin secretion (GSIS). However, the roles of CRFR1 in energy metabolism beyond insulin release remain elusive. MATERIALS/METHODS We characterized the metabolic phenotypes of mice lacking CRFR1 (CRFR1KO mice) under conditions of energy excess. RESULTS When fed a normal diet, the glucose profile of CRFR1KO mice in response to a glucose tolerance test was similar to that of wild-type (WT) mice, while serum insulin levels were significantly lower in CRFR1KO mice, reflecting high insulin sensitivity in part due to very low glucocorticoid levels. Histology of the pancreas revealed islet hypoplasia in CRFR1KO mice, suggesting a role of CRFR1 in maintaining the β cell mass in a manner similar to incretins. In response to a high-fat diet, CRFR1KO mice showed insulin resistance, but serum insulin levels during glucose challenge remained at a low level, indicating defective GSIS. In addition, CRFR1KO mice showed resistance to diet-induced obesity and hepatic steatosis. Although total food intake was not different between CRFR1KO and WT mice, oxygen consumption was significantly increased in CRFR1KO mice. The increased energy expenditure may explain the lean phenotype of CRFR1KO mice under conditions of energy excess. CONCLUSIONS Our results suggest that CRFR1 plays important roles in whole body energy homeostasis, providing compelling evidence of the close relationship between energy homeostasis and the function of the hypothalamic-pituitary-adrenal axis.
Collapse
Affiliation(s)
- Ryuichi Sakamoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Science, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Squillacioti C, De Luca A, Alì S, Ciarcia R, Germano G, Vittoria A, Mirabella N. Presence and distribution of urocortin and corticotrophin-releasing hormone receptors in the bovine thyroid gland. Anat Histol Embryol 2013; 43:429-34. [PMID: 24219151 DOI: 10.1111/ahe.12094] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 10/06/2013] [Indexed: 11/28/2022]
Abstract
Urocortin (UCN), a 40 amino acid peptide, is a corticotrophin-releasing hormone (CRH)-related peptide. The biological actions of CRH family peptides are mediated via two types of G-protein-coupled receptors, CRH type 1 (CRHR1) and CRH type 2 (CRHR2). The aim of this study was to investigate the expression of UCN, CRHR1 and CRHR2 by immunoprecipitation, Western blot, immunohistochemistry and RT-PCR in the bovine thyroid gland. Immunoprecipitation and Western blot analysis showed that tissue extracts reacted with the anti-UCN, anti-CRHR1 and anti-CRHR2 antibodies. RT-PCR experiments demonstrated that mRNAs of UCN, CRHR1 and CRHR2 were expressed. UCN immunoreactivity (IR) and CRHR2-IR were found in the thyroid follicular and parafollicular cells and CRHR1-IR in the smooth muscle of the blood vessels. These results suggest that a regulatory system exists in the bovine thyroid gland based on UCN, CRHR1 and CRHR2 and that UCN plays a role in the regulation of thyroid physiological functions through an autocrine/paracrine mechanism.
Collapse
Affiliation(s)
- C Squillacioti
- Dipartimento di Medicina Veterinaria e Produzioni animali, Università di Napoli "Federico II", Via Federico Delpino, 1-80137, Napoli, Italia
| | | | | | | | | | | | | |
Collapse
|
44
|
Amisten S, Salehi A, Rorsman P, Jones PM, Persaud SJ. An atlas and functional analysis of G-protein coupled receptors in human islets of Langerhans. Pharmacol Ther 2013; 139:359-91. [PMID: 23694765 DOI: 10.1016/j.pharmthera.2013.05.004] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 05/03/2013] [Indexed: 12/17/2022]
Abstract
G-protein coupled receptors (GPCRs) regulate hormone secretion from islets of Langerhans, and recently developed therapies for type-2 diabetes target islet GLP-1 receptors. However, the total number of GPCRs expressed by human islets, as well as their function and interactions with drugs, is poorly understood. In this review we have constructed an atlas of all GPCRs expressed by human islets: the 'islet GPCRome'. We have used this atlas to describe how islet GPCRs interact with their endogenous ligands, regulate islet hormone secretion, and interact with drugs known to target GPCRs, with a focus on drug/receptor interactions that may affect insulin secretion. The islet GPCRome consists of 293 GPCRs, a majority of which have unknown effects on insulin, glucagon and somatostatin secretion. The islet GPCRs are activated by 271 different endogenous ligands, at least 131 of which are present in islet cells. A large signalling redundancy was also found, with 119 ligands activating more than one islet receptor. Islet GPCRs are also the targets of a large number of clinically used drugs, and based on their coupling characteristics and effects on receptor signalling we identified 107 drugs predicted to stimulate and 184 drugs predicted to inhibit insulin secretion. The islet GPCRome highlights knowledge gaps in the current understanding of islet GPCR function, and identifies GPCR/ligand/drug interactions that might affect insulin secretion, which are important for understanding the metabolic side effects of drugs. This approach may aid in the design of new safer therapeutic agents with fewer detrimental effects on islet hormone secretion.
Collapse
Affiliation(s)
- Stefan Amisten
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, King's College London School of Medicine, London, UK.
| | | | | | | | | |
Collapse
|
45
|
Kubat E, Mahajan S, Liao M, Ackerman L, Ohara PT, Grady EF, Bhargava A. Corticotropin-releasing factor receptor 2 mediates sex-specific cellular stress responses. Mol Med 2013; 19:212-22. [PMID: 23835907 DOI: 10.2119/molmed.2013.00036] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 07/01/2013] [Indexed: 01/11/2023] Open
Abstract
Although females suffer twice as much as males from stress-related disorders, sex-specific participating and pathogenic cellular stress mechanisms remain uncharacterized. Using corticotropin-releasing factor receptor 2-deficient (Crhr2-/-) and wild-type (WT) mice, we show that CRF receptor type 2 (CRF2) and its high-affinity ligand, urocortin 1 (Ucn1), are key mediators of the endoplasmic reticulum (ER) stress response in a murine model of acute pancreatic inflammation. Ucn1 was expressed de novo in acinar cells of male, but not female WT mice during acute inflammation. Upon insult, acinar Ucn1 induction was markedly attenuated in male but not female Crhr2-/- mice. Crhr2-/- mice of both sexes show exacerbated acinar cell inflammation and necrosis. Electron microscopy showed mild ER damage in WT male mice and markedly distorted ER structure in Crhr2-/- male mice during pancreatitis. WT and Crhr2-/- female mice showed similarly distorted ER ultrastructure that was less severe than distortion seen in Crhr2-/- male mice. Damage in ER structure was accompanied by increased ubiquitination, peIF2, and mistargeted localization of vimentin in WT mice that was further exacerbated in Crhr2-/- mice of both sexes during pancreatitis. Exogenous Ucn1 rescued many aspects of histological damage and cellular stress response, including restoration of ER structure in male WT and Crhr2-/- mice, but not in females. Instead, females often showed increased damage. Thus, specific cellular pathways involved in coping and resolution seem to be distinct to each sex. Our results demonstrate the importance of identifying sex-specific pathogenic mechanisms and their value in designing effective therapeutics.
Collapse
Affiliation(s)
- Eric Kubat
- Department of Surgery, University of California San Francisco, San Francisco, California 94143, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Jensen MV, Haldeman JM, Zhang H, Lu D, Huising MO, Vale WW, Hohmeier HE, Rosenberg P, Newgard CB. Control of voltage-gated potassium channel Kv2.2 expression by pyruvate-isocitrate cycling regulates glucose-stimulated insulin secretion. J Biol Chem 2013; 288:23128-40. [PMID: 23788641 DOI: 10.1074/jbc.m113.491654] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Recent studies have shown that the pyruvate-isocitrate cycling pathway, involving the mitochondrial citrate/isocitrate carrier and the cytosolic NADP-dependent isocitrate dehydrogenase (ICDc), is involved in control of glucose-stimulated insulin secretion (GSIS). Here we demonstrate that pyruvate-isocitrate cycling regulates expression of the voltage-gated potassium channel family member Kv2.2 in islet β-cells. siRNA-mediated suppression of ICDc, citrate/isocitrate carrier, or Kv2.2 expression impaired GSIS, and the effect of ICDc knockdown was rescued by re-expression of Kv2.2. Moreover, chronic exposure of β-cells to elevated fatty acids, which impairs GSIS, resulted in decreased expression of Kv2.2. Surprisingly, knockdown of ICDc or Kv2.2 increased rather than decreased outward K(+) current in the 832/13 β-cell line. Immunoprecipitation studies demonstrated interaction of Kv2.1 and Kv2.2, and co-overexpression of the two channels reduced outward K(+) current compared with overexpression of Kv2.1 alone. Also, siRNA-mediated knockdown of ICDc enhanced the suppressive effect of the Kv2.1-selective inhibitor stromatoxin1 on K(+) currents. Our data support a model in which a key function of the pyruvate-isocitrate cycle is to maintain levels of Kv2.2 expression sufficient to allow it to serve as a negative regulator of Kv channel activity.
Collapse
Affiliation(s)
- Mette V Jensen
- Duke Institute of Molecular Physiology, Duke University Medical Center, Durham, North Carolina 27704, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Research status and prospect of stem cells in the treatment of diabetes mellitus. SCIENCE CHINA-LIFE SCIENCES 2013; 56:306-12. [DOI: 10.1007/s11427-013-4469-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 03/08/2013] [Indexed: 02/06/2023]
|
48
|
Abstract
Neurogenesis during embryonic and adult life is tightly regulated by a network of transcriptional, growth and hormonal factors. Emerging evidence indicates that activation of the stress response, via the associated glucocorticoid increase, reduces neurogenesis and contributes to the development of adult diseases.As corticotrophin-releasing hormone (CRH) or factor is the major mediator of adaptive response to stressors, we sought to investigate its involvement in this process. Accordingly, we found that CRH could reverse the damaging effects of glucocorticoid on neural stem/progenitor cells (NS/PCs), while its genetic deficiency results in compromised proliferation and enhanced apoptosis during neurogenesis. Analyses in fetal and adult mouse brain revealed significant expression of CRH receptors in proliferating neuronal progenitors. Furthermore, by using primary cultures of NS/PCs, we characterized the molecular mechanisms and identified CRH receptor-1 as the receptor mediating the neuroprotective effects of CRH. Finally, we demonstrate the expression of CRH receptors in human fetal brain from early gestational age, in areas of active neuronal proliferation. These observations raise the intriguing possibility for CRH-mediated pharmacological applications in diseases characterized by altered neuronal homeostasis, including depression, dementia, neurodegenerative diseases, brain traumas and obesity.
Collapse
|
49
|
Bonfiglio JJ, Inda C, Senin S, Maccarrone G, Refojo D, Giacomini D, Turck CW, Holsboer F, Arzt E, Silberstein S. B-Raf and CRHR1 internalization mediate biphasic ERK1/2 activation by CRH in hippocampal HT22 Cells. Mol Endocrinol 2013; 27:491-510. [PMID: 23371389 DOI: 10.1210/me.2012-1359] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
CRH is a key regulator of neuroendocrine, autonomic, and behavioral response to stress. CRH-stimulated CRH receptor 1 (CRHR1) activates ERK1/2 depending on intracellular context. In a previous work, we demonstrated that CRH activates ERK1/2 in limbic areas of the mouse brain (hippocampus and basolateral amygdala). ERK1/2 is an essential mediator of hippocampal physiological processes including emotional behavior, synaptic plasticity, learning, and memory. To elucidate the molecular mechanisms by which CRH activates ERK1/2 in hippocampal neurons, we used the mouse hippocampal cell line HT22. We document for the first time that ERK1/2 activation in response to CRH is biphasic, involving a first cAMP- and B-Raf-dependent early phase and a second phase that critically depends on CRHR1 internalization and β-arrestin2. By means of mass-spectrometry-based screening, we identified B-Raf-associated proteins that coimmunoprecipitate with endogenous B-Raf after CRHR1 activation. Using molecular and pharmacological tools, the functional impact of selected B-Raf partners in CRH-dependent ERK1/2 activation was dissected. These results indicate that 14-3-3 proteins, protein kinase A, and Rap1, are essential for early CRH-induced ERK1/2 activation, whereas dynamin and vimentin are required for the CRHR1 internalization-dependent phase. Both phases of ERK1/2 activation depend on calcium influx and are affected by calcium/calmodulin-dependent protein kinase II inactivation. Thus, this report describes the dynamics and biphasic nature of ERK1/2 activation downstream neuronal CRHR1 and identifies several new critical components of the CRHR1 signaling machinery that selectively controls the early and late phases of ERK1/2 activation, thus providing new potential therapeutic targets for stress-related disorders.
Collapse
Affiliation(s)
- Juan J Bonfiglio
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), CONICET, Partner Institute of the Max Planck Society, Godoy Cruz 2390, C1425FQA Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
van der Meulen T, Xie R, Kelly OG, Vale WW, Sander M, Huising MO. Urocortin 3 marks mature human primary and embryonic stem cell-derived pancreatic alpha and beta cells. PLoS One 2012; 7:e52181. [PMID: 23251699 PMCID: PMC3522648 DOI: 10.1371/journal.pone.0052181] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 11/16/2012] [Indexed: 12/28/2022] Open
Abstract
The peptide hormone Urocortin 3 (Ucn 3) is abundantly and exclusively expressed in mouse pancreatic beta cells where it regulates insulin secretion. Here we demonstrate that Ucn 3 first appears at embryonic day (E) 17.5 and, from approximately postnatal day (p) 7 and onwards throughout adult life, becomes a unifying and exclusive feature of mouse beta cells. These observations identify Ucn 3 as a potential beta cell maturation marker. To determine whether Ucn 3 is similarly restricted to beta cells in humans, we conducted comprehensive immunohistochemistry and gene expression experiments on macaque and human pancreas and sorted primary human islet cells. This revealed that Ucn 3 is not restricted to the beta cell lineage in primates, but is also expressed in alpha cells. To substantiate these findings, we analyzed human embryonic stem cell (hESC)-derived pancreatic endoderm that differentiates into mature endocrine cells upon engraftment in mice. Ucn 3 expression in hESC-derived grafts increased robustly upon differentiation into mature endocrine cells and localized to both alpha and beta cells. Collectively, these observations confirm that Ucn 3 is expressed in adult beta cells in both mouse and human and appears late in beta cell differentiation. Expression of Pdx1, Nkx6.1 and PC1/3 in hESC-derived Ucn 3+ beta cells supports this. However, the expression of Ucn 3 in primary and hESC-derived alpha cells demonstrates that human Ucn 3 is not exclusive to the beta cell lineage but is a general marker for both the alpha and beta cell lineages. Ucn 3+ hESC-derived alpha cells do not express Nkx6.1, Pdx1 or PC1/3 in agreement with the presence of a separate population of Ucn 3+ alpha cells. Our study highlights important species differences in Ucn 3 expression, which have implications for its utility as a marker to identify mature beta cells in (re)programming strategies.
Collapse
Affiliation(s)
- Talitha van der Meulen
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Ruiyu Xie
- Department of Pediatrics and Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Olivia G. Kelly
- ViaCyte, Inc., San Diego, California, United States of America
| | - Wylie W. Vale
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Maike Sander
- Department of Pediatrics and Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Mark O. Huising
- Peptide Biology Laboratories, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Salk Center for Nutritional Genomics, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|