1
|
Wang Y, Zhang JW, Wang JW, Wang JL, Zhang SC, Ma RY, Zhang J, Li Y, Liu PJ, Xue WJ, Zheng J, Ding XM. BMSCs overexpressed ISL1 reduces the apoptosis of islet cells through ANLN carrying exosome, INHBA, and caffeine. Cell Mol Life Sci 2022; 79:538. [PMID: 36190571 PMCID: PMC11802980 DOI: 10.1007/s00018-022-04571-0] [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: 06/05/2022] [Revised: 08/28/2022] [Accepted: 09/22/2022] [Indexed: 11/24/2022]
Abstract
Early apoptosis of grafted islets is one of the main factors affecting the efficacy of islet transplantation. The combined transplantation of islet cells and bone marrow mesenchymal stem cells (BMSCs) can significantly improve the survival rate of grafted islets. Transcription factor insulin gene enhancer binding protein 1 (ISL1) is shown to promote the angiogenesis of grafted islets and the paracrine function of mesenchymal stem cells during the co-transplantation, yet the regulatory mechanism remains unclear. By using ISL1-overexpressing BMSCs and the subtherapeutic doses of islets for co-transplantation, we managed to reduce the apoptosis and improve the survival rate of the grafts. Our metabolomics and proteomics data suggested that ISL1 upregulates aniline (ANLN) and Inhibin beta A chain (INHBA), and stimulated the release of caffeine in the BMSCs. We then demonstrated that the upregulation of ANLN and INHBA was achieved by the binding of ISL1 to the promoter regions of the two genes. In addition, ISL1 could also promote BMSCs to release exosomes with high expression of ANLN, secrete INHBA and caffeine, and reduce streptozocin (STZ)-induced islets apoptosis. Thus, our study provides mechanical insight into the islet/BMSCs co-transplantation and paves the foundation for using conditioned medium to mimic the ISL1-overexpressing BMSCs co-transplantation.
Collapse
Affiliation(s)
- Ying Wang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an 710061, Shaanxi, China
| | - Jiang-Wei Zhang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an 710061, Shaanxi, China
| | - Jing-Wen Wang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an 710061, Shaanxi, China
| | - Jia-Le Wang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an 710061, Shaanxi, China
| | - Shu-Cong Zhang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an 710061, Shaanxi, China
| | - Rui-Yang Ma
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an 710061, Shaanxi, China
| | - Jing Zhang
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an 710061, Shaanxi, China
| | - Yang Li
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an 710061, Shaanxi, China
| | - Pei-Jun Liu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi, China
| | - Wu-Jun Xue
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an 710061, Shaanxi, China
| | - Jin Zheng
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an 710061, Shaanxi, China
| | - Xiao-Ming Ding
- Department of Renal Transplantation, Hospital of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an 710061, Shaanxi, China.
| |
Collapse
|
2
|
Brown ML, Lopez A, Meyer N, Richter A, Thompson TB. FSTL3-Neutralizing Antibodies Enhance Glucose-Responsive Insulin Secretion in Dysfunctional Male Mouse and Human Islets. Endocrinology 2021; 162:6128796. [PMID: 33539535 PMCID: PMC8384134 DOI: 10.1210/endocr/bqab019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Indexed: 12/23/2022]
Abstract
Diabetes is caused by insufficient insulin production from pancreatic beta cells or insufficient insulin action, leading to an inability to control blood glucose. While a wide range of treatments exist to alleviate the symptoms of diabetes, therapies addressing the root cause of diabetes through replacing lost beta cells with functional cells remain an object of active pursuit. We previously demonstrated that genetic deletion of Fstl3, a critical regulator of activin activity, enhanced beta cell number and glucose-responsive insulin production. These observations suggested the hypothesis that FSTL3 neutralization could be used to therapeutically enhance beta cell number and function in humans. To pursue this possibility, we developed an FSTL3-neutralizing antibody, FP-101, and characterized its ability to prevent or disrupt FSTL3 from complexing with activin or related ligands. This antibody was selective for FSTL3 relative to the closely related follistatin, thereby reducing the chance for off-target effects. In vitro assays with FP-101 and activin revealed that FP-101-mediated neutralization of FSTL3 can enhance both insulin secretion and glucose responsiveness to nonfunctional mouse and human islets under conditions that model diabetes. Thus, FSTL3 neutralization may provide a novel therapeutic strategy for treating diabetes through repairing dysfunctional beta cells.
Collapse
Affiliation(s)
- Melissa L Brown
- Department of Nutrition and Public Health, University of Saint Joseph, West Hartford, CT 06117, USA
- Correspondence: Melissa Brown, PhD, RD, CSSD, LD, University of Saint Joseph, 1678 Asylum Ave, West Hartford, CT 06117, USA. E-mail:
| | - Alexa Lopez
- Fairbanks Pharmaceuticals, Inc., Concord, MA 01742, USA
| | - Nolan Meyer
- Fairbanks Pharmaceuticals, Inc., Concord, MA 01742, USA
| | - Alden Richter
- Fairbanks Pharmaceuticals, Inc., Concord, MA 01742, USA
| | - Thomas B Thompson
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
3
|
Brown ML, Schneyer A. A Decade Later: Revisiting the TGFβ Family's Role in Diabetes. Trends Endocrinol Metab 2021; 32:36-47. [PMID: 33261990 DOI: 10.1016/j.tem.2020.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/16/2022]
Abstract
In 2010, we published a review summarizing the role of the transforming growth factor-beta (TGFβ) family of proteins in diabetes. At that time there were still many outstanding questions that needed to be answered. In this updated review, we revisit the topic and provide new evidence that supports findings from previous studies included in the 2010 review and adds to the knowledge base with new findings and information. The most substantial contributions in the past 10 years have been in the areas of human data, the investigation of TGFβ family members other than activin [e.g., bone morphogenetic proteins (BMPs), growth and differentiation factor 11 (GDF11), nodal], and the expansion of β-cell number through various mechanisms including transdifferentiation, which was previously believed to not be possible.
Collapse
Affiliation(s)
| | - Alan Schneyer
- Fairbanks Pharmaceuticals, Inc., Springfield, MA 01199, USA
| |
Collapse
|
4
|
Sakhneny L, Rachi E, Epshtein A, Guez HC, Wald-Altman S, Lisnyansky M, Khalifa-Malka L, Hazan A, Baer D, Priel A, Weil M, Landsman L. Pancreatic Pericytes Support β-Cell Function in a Tcf7l2-Dependent Manner. Diabetes 2018; 67:437-447. [PMID: 29246974 DOI: 10.2337/db17-0697] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 12/06/2017] [Indexed: 01/13/2023]
Abstract
Polymorphism in TCF7L2, a component of the canonical Wnt signaling pathway, has a strong association with β-cell dysfunction and type 2 diabetes through a mechanism that has yet to be defined. β-Cells rely on cells in their microenvironment, including pericytes, for their proper function. Here, we show that Tcf7l2 activity in pancreatic pericytes is required for β-cell function. Transgenic mice in which Tcf7l2 was selectively inactivated in their pancreatic pericytes exhibited impaired glucose tolerance due to compromised β-cell function and glucose-stimulated insulin secretion. Inactivation of pericytic Tcf7l2 was associated with impaired expression of genes required for β-cell function and maturity in isolated islets. In addition, we identified Tcf7l2-dependent pericytic expression of secreted factors shown to promote β-cell function, including bone morphogenetic protein 4 (BMP4). Finally, we show that exogenous BMP4 is sufficient to rescue the impaired glucose-stimulated insulin secretion of transgenic mice, pointing to a potential mechanism through which pericytic Tcf7l2 activity affects β-cells. To conclude, we suggest that pancreatic pericytes produce secreted factors, including BMP4, in a Tcf7l2-dependent manner to support β-cell function. Our findings thus propose a potential cellular mechanism through which abnormal TCF7L2 activity predisposes individuals to diabetes and implicates abnormalities in the islet microenvironment in this disease.
Collapse
Affiliation(s)
- Lina Sakhneny
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eleonor Rachi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alona Epshtein
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Helen C Guez
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shane Wald-Altman
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences and the Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Michal Lisnyansky
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Laura Khalifa-Malka
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adina Hazan
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Daria Baer
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avi Priel
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Miguel Weil
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences and the Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Limor Landsman
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
5
|
Li H, Li Y, Xiang L, Zhang J, Zhu B, Xiang L, Dong J, Liu M, Xiang G. GDF11 Attenuates Development of Type 2 Diabetes via Improvement of Islet β-Cell Function and Survival. Diabetes 2017; 66:1914-1927. [PMID: 28450417 DOI: 10.2337/db17-0086] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/18/2017] [Indexed: 11/13/2022]
Abstract
Growth differentiation factor 11 (GDF11) has been implicated in the regulation of islet development and a variety of aging conditions, but little is known about the physiological functions of GDF11 in adult pancreatic islets. Here, we showed that systematic replenishment of GDF11 not only preserved insulin secretion but also improved the survival and morphology of β-cells and improved glucose metabolism in both nongenetic and genetic mouse models of type 2 diabetes (T2D). Conversely, anti-GDF11 monoclonal antibody treatment caused β-cell failure and lethal T2D. In vitro treatment of isolated murine islets and MIN6 cells with recombinant GDF11 attenuated glucotoxicity-induced β-cell dysfunction and apoptosis. Mechanistically, the GDF11-mediated protective effects could be attributed to the activation of transforming growth factor-β/Smad2 and phosphatidylinositol-4,5-bisphosphate 3-kinase-AKT-FoxO1 signaling. These findings suggest that GDF11 repletion may improve β-cell function and mass and thus may lead to a new therapeutic approach for T2D.
Collapse
Affiliation(s)
- Huan Li
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| | - Yixiang Li
- Radiation-Diagnostic/Oncology School of Medicine, Emory University, Atlanta, GA
| | - Lingwei Xiang
- Mathematics and Statistics Department, Georgia State University, Atlanta, GA
| | - JiaJia Zhang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| | - Biao Zhu
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| | - Lin Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| | - Jing Dong
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| | - Min Liu
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| | - Guangda Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei Province, China
| |
Collapse
|
6
|
Maresch CC, Stute DC, Ludlow H, Hammes HP, de Kretser DM, Hedger MP, Linn T. Hyperglycemia is associated with reduced testicular function and activin dysregulation in the Ins2 Akita+/- mouse model of type 1 diabetes. Mol Cell Endocrinol 2017; 446:91-101. [PMID: 28214591 DOI: 10.1016/j.mce.2017.02.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 02/09/2017] [Accepted: 02/12/2017] [Indexed: 01/23/2023]
Abstract
Type 1 diabetes (T1D) is associated with subfertility in men. We hypothesised that this results from inhibitory effects of chronic hyperglycemia on testicular function and used the Ins2Akita+/- mouse model to investigate this. Diabetic mice exhibited progressive testicular dysfunction, with a 30% reduction in testis weight at 24 weeks of age. Diabetic mice showed significantly reduced seminiferous tubule diameters and increased spermatogenic disruption, although testes morphology appeared grossly normal. Unexpectedly, serum LH and intra-testicular testosterone were similar in all groups. Ins2Akita+/- mice displayed elevation of the testicular inflammatory cytokines activin A and IL-6. Intratesticular activin B was downregulated, while the activin regulatory proteins, follistatin and inhibin, were unchanged. Activin signalling, measured by pSmad3 and Smad4 production, was enhanced in diabetic mice only. These results suggest that prolonged exposure to hyperglycemia in the Ins2Akita+/- mice leads to progressive testicular disruption mediated by testicular activin activity, rather than hormonal dysregulation.
Collapse
Affiliation(s)
- Constanze C Maresch
- Clinical Research Unit, Centre of Internal Medicine, Justus-Liebig-University, Giessen, Germany; Hudson Institute of Medical Research and Department of Anatomy & Developmental Biology, Monash University, Melbourne, Australia.
| | - Dina C Stute
- Clinical Research Unit, Centre of Internal Medicine, Justus-Liebig-University, Giessen, Germany
| | | | - Hans-Peter Hammes
- V. Medical Dept., Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - David M de Kretser
- Hudson Institute of Medical Research and Department of Anatomy & Developmental Biology, Monash University, Melbourne, Australia
| | - Mark P Hedger
- Hudson Institute of Medical Research and Department of Anatomy & Developmental Biology, Monash University, Melbourne, Australia
| | - Thomas Linn
- Clinical Research Unit, Centre of Internal Medicine, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
7
|
Namwanje M, Brown CW. Activins and Inhibins: Roles in Development, Physiology, and Disease. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a021881. [PMID: 27328872 DOI: 10.1101/cshperspect.a021881] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Since their original discovery as regulators of follicle-stimulating hormone (FSH) secretion and erythropoiesis, the TGF-β family members activin and inhibin have been shown to participate in a variety of biological processes, from the earliest stages of embryonic development to highly specialized functions in terminally differentiated cells and tissues. Herein, we present the history, structures, signaling mechanisms, regulation, and biological processes in which activins and inhibins participate, including several recently discovered biological activities and functional antagonists. The potential therapeutic relevance of these advances is also discussed.
Collapse
Affiliation(s)
- Maria Namwanje
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Chester W Brown
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030 Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030 Texas Children's Hospital, Houston, Texas 77030
| |
Collapse
|
8
|
Xiao X, Fischbach S, Song Z, Gaffar I, Zimmerman R, Wiersch J, Prasadan K, Shiota C, Guo P, Ramachandran S, Witkowski P, Gittes GK. Transient Suppression of TGFβ Receptor Signaling Facilitates Human Islet Transplantation. Endocrinology 2016; 157:1348-56. [PMID: 26872091 PMCID: PMC4816736 DOI: 10.1210/en.2015-1986] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although islet transplantation is an effective treatment for severe diabetes, its broad application is greatly limited due to a shortage of donor islets. Suppression of TGFβ receptor signaling in β-cells has been shown to increase β-cell proliferation in mice, but has not been rigorously examined in humans. Here, treatment of human islets with a TGFβ receptor I inhibitor, SB-431542 (SB), significantly improved C-peptide secretion by β-cells, and significantly increased β-cell number by increasing β-cell proliferation. In addition, SB increased cell-cycle activators and decreased cell-cycle suppressors in human β-cells. Transplantation of SB-treated human islets into diabetic immune-deficient mice resulted in significant improvement in blood glucose control, significantly higher serum and graft insulin content, and significantly greater increases in β-cell proliferation in the graft, compared with controls. Thus, our data suggest that transient suppression of TGFβ receptor signaling may improve the outcome of human islet transplantation, seemingly through increasing β-cell number and function.
Collapse
|
9
|
Brown ML, Andrzejewski D, Burnside A, Schneyer AL. Activin Enhances α- to β-Cell Transdifferentiation as a Source For β-Cells In Male FSTL3 Knockout Mice. Endocrinology 2016; 157:1043-54. [PMID: 26727106 DOI: 10.1210/en.2015-1793] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Diabetes results from inadequate β-cell number and/or function to control serum glucose concentrations so that replacement of lost β-cells could become a viable therapy for diabetes. In addition to embryonic stem cell sources for new β-cells, evidence for transdifferentiation/reprogramming of non-β-cells to functional β-cells is accumulating. In addition, de-differentiation of β-cells observed in diabetes and their subsequent conversion to α-cells raises the possibility that adult islet cell fate is malleable and controlled by local hormonal and/or environmental cues. We previously demonstrated that inactivation of the activin antagonist, follistatin-like 3 (FSTL3) resulted in β-cell expansion and improved glucose homeostasis in the absence of β-cell proliferation. We recently reported that activin directly suppressed expression of critical α-cell genes while increasing expression of β-cell genes, supporting the hypothesis that activin is one of the local hormones controlling islet cell fate and that increased activin signaling accelerates α- to β-cell transdifferentiation. We tested this hypothesis using Gluc-Cre/yellow fluorescent protein (YFP) α-cell lineage tracing technology combined with FSTL3 knockout (KO) mice to label α-cells with YFP. Flow cytometry was used to quantify unlabeled and labeled α- and β-cells. We found that Ins+/YFP+ cells were significantly increased in FSTL3 KO mice compared with wild type littermates. Labeled Ins+/YFP+ cells increased significantly with age in FSTL3 KO mice but not wild type littermates. Sorting results were substantiated by counting fluorescently labeled cells in pancreatic sections. Activin treatment of isolated islets significantly increased the number of YFP+/Ins+ cells. These results suggest that α- to β-cell transdifferentiation is influenced by activin signaling and may contribute substantially to β-cell mass.
Collapse
Affiliation(s)
- Melissa L Brown
- Departments of Nutrition (M.L.B.) and Veterinary and Animal Science (D.A., A.B., A.L.S.), University of Massachusetts-Amherst, Amherst, Massachusetts 01003
| | - Danielle Andrzejewski
- Departments of Nutrition (M.L.B.) and Veterinary and Animal Science (D.A., A.B., A.L.S.), University of Massachusetts-Amherst, Amherst, Massachusetts 01003
| | - Amy Burnside
- Departments of Nutrition (M.L.B.) and Veterinary and Animal Science (D.A., A.B., A.L.S.), University of Massachusetts-Amherst, Amherst, Massachusetts 01003
| | - Alan L Schneyer
- Departments of Nutrition (M.L.B.) and Veterinary and Animal Science (D.A., A.B., A.L.S.), University of Massachusetts-Amherst, Amherst, Massachusetts 01003
| |
Collapse
|
10
|
ActivinB Is Induced in Insulinoma To Promote Tumor Plasticity through a β-Cell-Induced Dedifferentiation. Mol Cell Biol 2015; 36:756-64. [PMID: 26711255 DOI: 10.1128/mcb.00930-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 12/13/2015] [Indexed: 12/19/2022] Open
Abstract
Loss of pancreatic β-cell maturity occurs in diabetes and insulinomas. Although both physiological and pathological stresses are known to promote β-cell dedifferentiation, little is known about the molecules involved in this process. Here we demonstrate that activinB, a transforming growth factor β (TGF-β)-related ligand, is upregulated during tumorigenesis and drives the loss of insulin expression and β-cell maturity in a mouse insulinoma model. Our data further identify Pax4 as a previously unknown activinB target and potent contributor to the observed β-cell dedifferentiation. More importantly, using compound mutant mice, we found that deleting activinB expression abolishes tumor β-cell dedifferentiation and, surprisingly, increases survival without significantly affecting tumor growth. Hence, this work reveals an unexpected role for activinB in the loss of β-cell maturity, islet plasticity, and progression of insulinoma through its participation in β-cell dedifferentiation.
Collapse
|
11
|
Jiang FX, Mishina Y, Baten A, Morahan G, Harrison LC. Transcriptome of pancreas-specific Bmpr1a-deleted islets links to TPH1-5-HT axis. Biol Open 2015; 4:1016-23. [PMID: 26187948 PMCID: PMC4542282 DOI: 10.1242/bio.011858] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Bone morphogenetic protein (BMP) signaling is crucial for the development and function of numerous organs, but its role on the function of pancreatic islets is not completely clear. To explore this question, we applied the high throughput transcriptomic analyses on the islets isolated from mice with a pancreas-specific deletion of the gene, Bmpr1a, encoding the type 1a BMP receptor. Consistently, these pBmpr1aKO mice had impaired glucose homeostasis at 3 months, and were more severely affected at 12 months of age. These had lower fasting blood insulin concentrations, with reduced expression of several key regulators of β-cell function. Importantly, transcriptomic profiling of 3-month pBmpr1aKO islets and bioinformatic analyses revealed abnormal expression of 203 metabolic genes. Critically among these, the tryptophan hydroxylase 1 gene (Tph1), encoding the rate-limiting enzyme for the production of 5-hydroxytryptamine (5-HT) was the highest over-expressed one. 5-HT is an important regulator of insulin secretion from β cells. Treatment with excess 5-HT inhibited this secretion. Thus our transcriptomic analysis links two highly conserved molecular pathways the BMP signaling and the TPH1–5-HT axis on glucose homeostasis.
Collapse
Affiliation(s)
- Fang-Xu Jiang
- The Walter & Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Akma Baten
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Grant Morahan
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, Western Australia 6009, Australia
| | - Leonard C Harrison
- The Walter & Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia
| |
Collapse
|
12
|
Andrzejewski D, Brown ML, Ungerleider N, Burnside A, Schneyer AL. Activins A and B Regulate Fate-Determining Gene Expression in Islet Cell Lines and Islet Cells From Male Mice. Endocrinology 2015; 156:2440-50. [PMID: 25961841 DOI: 10.1210/en.2015-1167] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TGFβ superfamily ligands, receptors, and second messengers, including activins A and B, have been identified in pancreatic islets and proposed to have important roles regulating development, proliferation, and function. We previously demonstrated that Fstl3 (an antagonist of activin activity) null mice have larger islets with β-cell hyperplasia and improved glucose tolerance and insulin sensitivity in the absence of altered β-cell proliferation. This suggested the hypothesis that increased activin signaling influences β-cell expansion by destabilizing the α-cell phenotype and promoting transdifferentiation to β-cells. We tested the first part of this hypothesis by treating α- and β-cell lines and sorted mouse islet cells with activin and related ligands. Treatment of the αTC1-6 α cell line with activins A or B suppressed critical α-cell gene expression, including Arx, glucagon, and MafB while also enhancing β-cell gene expression. In INS-1E β-cells, activin A treatment induced a significant increase in Pax4 (a fate determining β-cell gene) and insulin expression. In sorted primary islet cells, α-cell gene expression was again suppressed by activin treatment in α-cells, whereas Pax4 was enhanced in β-cells. Activin treatment in both cell lines and primary cells resulted in phosphorylated mothers against decapentaplegic-2 phosphorylation. Finally, treatment of αTC1-6 cells with activins A or B significantly inhibited proliferation. These results support the hypothesis that activin signaling destabilized the α-cell phenotype while promoting a β-cell fate. Moreover, these results support a model in which the β-cell expansion observed in Fstl3 null mice may be due, at least in part, to enhanced α- to β-cell transdifferentiation.
Collapse
Affiliation(s)
- Danielle Andrzejewski
- Departments of Veterinary and Animal Science (D.A., A.B., A.L.S.) and Nutrition (M.L.B.), and Molecular and Cellular Biology Graduate Program (N.U.), University of Massachusetts-Amherst, Amherst, Massachusetts 01003
| | - Melissa L Brown
- Departments of Veterinary and Animal Science (D.A., A.B., A.L.S.) and Nutrition (M.L.B.), and Molecular and Cellular Biology Graduate Program (N.U.), University of Massachusetts-Amherst, Amherst, Massachusetts 01003
| | - Nathan Ungerleider
- Departments of Veterinary and Animal Science (D.A., A.B., A.L.S.) and Nutrition (M.L.B.), and Molecular and Cellular Biology Graduate Program (N.U.), University of Massachusetts-Amherst, Amherst, Massachusetts 01003
| | - Amy Burnside
- Departments of Veterinary and Animal Science (D.A., A.B., A.L.S.) and Nutrition (M.L.B.), and Molecular and Cellular Biology Graduate Program (N.U.), University of Massachusetts-Amherst, Amherst, Massachusetts 01003
| | - Alan L Schneyer
- Departments of Veterinary and Animal Science (D.A., A.B., A.L.S.) and Nutrition (M.L.B.), and Molecular and Cellular Biology Graduate Program (N.U.), University of Massachusetts-Amherst, Amherst, Massachusetts 01003
| |
Collapse
|
13
|
Stewart AF, Hussain MA, García-Ocaña A, Vasavada RC, Bhushan A, Bernal-Mizrachi E, Kulkarni RN. Human β-cell proliferation and intracellular signaling: part 3. Diabetes 2015; 64:1872-85. [PMID: 25999530 PMCID: PMC4439562 DOI: 10.2337/db14-1843] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This is the third in a series of Perspectives on intracellular signaling pathways coupled to proliferation in pancreatic β-cells. We contrast the large knowledge base in rodent β-cells with the more limited human database. With the increasing incidence of type 1 diabetes and the recognition that type 2 diabetes is also due in part to a deficiency of functioning β-cells, there is great urgency to identify therapeutic approaches to expand human β-cell numbers. Therapeutic approaches might include stem cell differentiation, transdifferentiation, or expansion of cadaver islets or residual endogenous β-cells. In these Perspectives, we focus on β-cell proliferation. Past Perspectives reviewed fundamental cell cycle regulation and its upstream regulation by insulin/IGF signaling via phosphatidylinositol-3 kinase/mammalian target of rapamycin signaling, glucose, glycogen synthase kinase-3 and liver kinase B1, protein kinase Cζ, calcium-calcineurin-nuclear factor of activated T cells, epidermal growth factor/platelet-derived growth factor family members, Wnt/β-catenin, leptin, and estrogen and progesterone. Here, we emphasize Janus kinase/signal transducers and activators of transcription, Ras/Raf/extracellular signal-related kinase, cadherins and integrins, G-protein-coupled receptors, and transforming growth factor β signaling. We hope these three Perspectives will serve to introduce these pathways to new researchers and will encourage additional investigators to focus on understanding how to harness key intracellular signaling pathways for therapeutic human β-cell regeneration for diabetes.
Collapse
Affiliation(s)
- Andrew F Stewart
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mehboob A Hussain
- Departments of Medicine and Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - Adolfo García-Ocaña
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rupangi C Vasavada
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Anil Bhushan
- Diabetes Center, University of California, San Francisco, San Francisco, CA
| | - Ernesto Bernal-Mizrachi
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan, Ann Arbor, MI, and VA Ann Arbor Healthcare System, Ann Arbor, MI
| | - Rohit N Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| |
Collapse
|
14
|
Zhao C, Qiao C, Tang RH, Jiang J, Li J, Martin CB, Bulaklak K, Li J, Wang DW, Xiao X. Overcoming Insulin Insufficiency by Forced Follistatin Expression in β-cells of db/db Mice. Mol Ther 2015; 23:866-874. [PMID: 25676679 PMCID: PMC4427879 DOI: 10.1038/mt.2015.29] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 02/04/2015] [Indexed: 12/19/2022] Open
Abstract
Diabetes poses a substantial burden to society as it can lead to serious complications and premature death. The number of cases continues to increase worldwide. Two major causes of diabetes are insulin resistance and insulin insufficiency. Currently, there are few antidiabetic drugs available that can preserve or protect β-cell function to overcome insulin insufficiency in diabetes. We describe a therapeutic strategy to preserve β-cell function by overexpression of follistatin (FST) using an AAV vector (AAV8-Ins-FST) in diabetic mouse model. Overexpression of FST in the pancreas of db/db mouse increased β-cell islet mass, decreased fasting glucose level, alleviated diabetic symptoms, and essentially doubled lifespan of the treated mice. The observed islet enlargement was attributed to β-cell proliferation as a result of bioneutralization of myostatin and activin by FST. Overall, our study indicates overexpression of FST in the diabetic pancreas preserves β-cell function by promoting β-cell proliferation, opening up a new therapeutic avenue for the treatment of diabetes.
Collapse
Affiliation(s)
- Chunxia Zhao
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA; Cardiovascular Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunping Qiao
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Ru-Hang Tang
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jiangang Jiang
- Cardiovascular Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianbin Li
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Carrie Bette Martin
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Karen Bulaklak
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Juan Li
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Dao Wen Wang
- Cardiovascular Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Xiao
- Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA.
| |
Collapse
|
15
|
Bruun C, Christensen GL, Jacobsen MLB, Kanstrup MB, Jensen PR, Fjordvang H, Mandrup-Poulsen T, Billestrup N. Inhibition of beta cell growth and function by bone morphogenetic proteins. Diabetologia 2014; 57:2546-54. [PMID: 25260823 DOI: 10.1007/s00125-014-3384-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/02/2014] [Indexed: 11/25/2022]
Abstract
AIMS/HYPOTHESIS Impairment of beta cell mass and function is evident in both type 1 and type 2 diabetes. In healthy physiological conditions pancreatic beta cells adapt to the body's increasing insulin requirements by proliferation and improved function. We hypothesised that during the development of diabetes, there is an increase in the expression of inhibitory factors that prevent the beta cells from adapting to the increased need for insulin. We evaluated the effects of bone morphogenetic protein (BMP) 2 and -4 on beta cells. METHODS The effects of BMP2 and -4 on beta cell proliferation, apoptosis, gene expression and insulin release were studied in isolated islets of Langerhans from rats, mice and humans. The expression of BMPs was analysed by immunocytochemistry and real-time PCR. The role of endogenous BMP was investigated using a soluble and neutralising form of the BMP receptor 1A. RESULTS BMP2 and -4 were found to inhibit basal as well as growth factor-stimulated proliferation of primary beta cells from rats and mice. Bmp2 and Bmp4 mRNA and protein were expressed in islets and regulated by inflammatory cytokines. Neutralisation of endogenous BMP activity resulted in enhanced proliferation of rodent beta cells. The expression of Id mRNAs was induced by BMP4 in rat and human islets. Finally, glucose-induced insulin secretion was significantly impaired in rodent and human islets pre-treated with BMP4, and inhibition of BMP activity resulted in enhanced insulin release. CONCLUSIONS/INTERPRETATION These data show that BMP2 and -4 exert inhibitory actions on beta cells in vitro and suggest that BMPs exert regulatory roles of beta cell growth and function.
Collapse
|
16
|
Wu H, Mezghenna K, Marmol P, Guo T, Moliner A, Yang SN, Berggren PO, Ibáñez CF. Differential regulation of mouse pancreatic islet insulin secretion and Smad proteins by activin ligands. Diabetologia 2014; 57:148-56. [PMID: 24132781 DOI: 10.1007/s00125-013-3079-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 09/23/2013] [Indexed: 10/26/2022]
Abstract
AIMS/HYPOTHESIS Glucose-stimulated insulin secretion (GSIS) from pancreatic beta cells is regulated by paracrine factors, the identity and mechanisms of action of which are incompletely understood. Activins are expressed in pancreatic islets and have been implicated in the regulation of GSIS. Activins A and B signal through a common set of intracellular components, but it is unclear whether they display similar or distinct functions in glucose homeostasis. METHODS We examined glucose homeostatic responses in mice lacking activin B and in pancreatic islets derived from these mutants. We compared the ability of activins A and B to regulate downstream signalling, ATP production and GSIS in islets and beta cells. RESULTS Mice lacking activin B displayed elevated serum insulin levels and GSIS. Injection of a soluble activin B antagonist phenocopied these changes in wild-type mice. Isolated pancreatic islets from mutant mice showed enhanced GSIS, which could be rescued by exogenous activin B. Activin B negatively regulated GSIS and ATP production in wild-type islets, while activin A displayed the opposite effects. The downstream mediator Smad3 responded preferentially to activin B in pancreatic islets and beta cells, while Smad2 showed a preference for activin A, indicating distinct signalling effects of the two activins. In line with this, overexpression of Smad3, but not Smad2, decreased GSIS in pancreatic islets. CONCLUSIONS/INTERPRETATION These results reveal a tug-of-war between activin ligands in the regulation of insulin secretion by beta cells, and suggest that manipulation of activin signalling could be a useful strategy for the control of glucose homeostasis in diabetes and metabolic disease.
Collapse
Affiliation(s)
- Haiya Wu
- Department of Neuroscience, Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Berzelius vag 35, B3 Box 285, SE-171 77, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Brown ML, Ungerleider N, Bonomi L, Andrzejewski D, Burnside A, Schneyer A. Effects of activin A on survival, function and gene expression of pancreatic islets from non-diabetic and diabetic human donors. Islets 2014; 6:e1017226. [PMID: 25833251 PMCID: PMC4398300 DOI: 10.1080/19382014.2015.1017226] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Emerging evidence suggests that activin with its associated receptors, second messengers, and antagonists would be excellent targets for therapeutic drug development in the treatment of diabetes. We undertook the current study to investigate the ability to extrapolate findings from rodent studies to human islets in which data thus far has been scarce. We tested the hypothesis that human islets synthesize activin and that activin participates in the regulation of islet β-cells. Human islets from 33 separate isolations were categorized based on functional status, culture status and diabetic status. Statistical comparisons were made by ANOVA with Tukey post-hoc adjustment for multiple comparisons. Experiments investigating activin utilized qPCR, FACS cell sorting, immunofluorescent antibody staining, functionality assays, viability assays and protein secretion assays. We have defined the transcript expression patterns of activin and the TGFβ superfamily in human islets. We found INHBA (the gene encoding activin A) to be the most highly expressed of the superfamily in normal, cultured islets. We elucidated a link between the islet microenvironment and activin A. We found differential ligand expression based on diabetic, culture and functional status. Further, this is also the first report that links direct effects of activin A with the ability to restore glucose-stimulated insulin secretion in human islets from type 2 diabetic donors thereby establishing the relevance of targeting activin for therapeutic drug development.
Collapse
Affiliation(s)
- Melissa L Brown
- University of Massachusetts
Amherst; Amherst, MA USA
- Pioneer Valley Life Sciences
Institute; Springfield, MA USA
- Department of Nutrition; University of
Massachusetts; Amherst, MA USA
- Correspondence to: Melissa L Brown;
| | - Nathan Ungerleider
- University of Massachusetts
Amherst; Amherst, MA USA
- Pioneer Valley Life Sciences
Institute; Springfield, MA USA
| | - Lara Bonomi
- University of Massachusetts
Amherst; Amherst, MA USA
- Pioneer Valley Life Sciences
Institute; Springfield, MA USA
| | - Danielle Andrzejewski
- University of Massachusetts
Amherst; Amherst, MA USA
- Pioneer Valley Life Sciences
Institute; Springfield, MA USA
- Department of Veterinary and Animal Sciences;
University of Massachusetts; Amherst, MA USA
| | - Amy Burnside
- University of Massachusetts
Amherst; Amherst, MA USA
- Pioneer Valley Life Sciences
Institute; Springfield, MA USA
- Department of Veterinary and Animal Sciences;
University of Massachusetts; Amherst, MA USA
| | - Alan Schneyer
- University of Massachusetts
Amherst; Amherst, MA USA
- Pioneer Valley Life Sciences
Institute; Springfield, MA USA
- Department of Veterinary and Animal Sciences;
University of Massachusetts; Amherst, MA USA
| |
Collapse
|
18
|
Gabillard JC, Biga PR, Rescan PY, Seiliez I. Revisiting the paradigm of myostatin in vertebrates: insights from fishes. Gen Comp Endocrinol 2013; 194:45-54. [PMID: 24018114 DOI: 10.1016/j.ygcen.2013.08.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 08/09/2013] [Accepted: 08/15/2013] [Indexed: 11/21/2022]
Abstract
In the last decade, myostatin (MSTN), a member of the TGFβ superfamily, has emerged as a strong inhibitor of muscle growth in mammals. In fish many studies reveal a strong conservation of mstn gene organization, sequence, and protein structures. Because of ancient genome duplication, teleostei may have retained two copies of mstn genes and even up to four copies in salmonids due to additional genome duplication event. In sharp contrast to mammals, the different fish mstn orthologs are widely expressed with a tissue-specific expression pattern. Quantification of mstn mRNA in fish under different physiological conditions, demonstrates that endogenous expression of mstn paralogs is rarely related to fish muscle growth rate. In addition, attempts to inhibit MSTN activity did not consistently enhance muscle growth as in mammals. In vitro, MSTN stimulates myotube atrophy and inhibits proliferation but not differentiation of myogenic cells as in mammals. In conclusion, given the strong mstn expression non-muscle tissues of fish, we propose a new hypothesis stating that fish MSTN functions as a general inhibitors of cell proliferation and cell growth to control tissue mass but is not specialized into a strong muscle regulator.
Collapse
Affiliation(s)
- Jean-Charles Gabillard
- INRA, UR1037 Laboratoire de Physiologie et Génomique des Poissons, Equipe Croissance et Qualité de la Chair des Poissons, Campus de Beaulieu, 35000 Rennes, France.
| | | | | | | |
Collapse
|
19
|
Szabat M, Johnson JD. Modulation of β-cell fate and function by TGFβ ligands: a superfamily with many powers. Endocrinology 2013; 154:3965-9. [PMID: 24141995 DOI: 10.1210/en.2013-1880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Marta Szabat
- PhD, Associate Professor, Medicine and Cellular and Physiological Sciences, Surgery, Diabetes Research Group, Cardiovascular Research Group, The University of British Columbia, Point Grey Campus, 5358-2350 Health Sciences Mall, Vancouver, British Columbia, Canada V6T 1Z3.
| | | |
Collapse
|
20
|
Vogel KJ, Brown MR, Strand MR. Phylogenetic investigation of Peptide hormone and growth factor receptors in five dipteran genomes. Front Endocrinol (Lausanne) 2013; 4:193. [PMID: 24379806 PMCID: PMC3863949 DOI: 10.3389/fendo.2013.00193] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 11/29/2013] [Indexed: 12/26/2022] Open
Abstract
Peptide hormones and growth factors bind to membrane receptors and regulate a myriad of processes in insects and other metazoans. The evolutionary relationships among characterized and uncharacterized ("orphan") receptors can provide insights into receptor-ligand biology and narrow target choices in deorphanization studies. However, the large number and low sequence conservation of these receptors make evolutionary analysis difficult. Here, we characterized the G-protein-coupled receptors (GPCRs), receptor guanylyl cyclases (RGCs), and protein kinase receptors (PKRs) of mosquitoes and select other flies by interrogating the genomes of Aedes aegypti, Anopheles gambiae, Culex quinquefasciatus, Drosophila melanogaster, and D. mojavensis. Sequences were grouped by receptor type, clustered using the program CLANS, aligned using HMMR, and phylogenetic trees built using PhyML. Our results indicated that PKRs had relatively few orphan clades whereas GPCRs and RGCs had several. In addition, more than half of the Class B secretin-like GPCRs and RGCs remained uncharacterized. Additional studies revealed that Class B GPCRs exhibited more gain and loss events than other receptor types. Finally, using the neuropeptide F family of insect receptors and the neuropeptide Y family of vertebrate receptors, we also show that functional sites considered critical for ligand binding are conserved among distinct family members and between distantly related taxa. Overall, our results provide the first comprehensive analysis of peptide hormone and growth factor receptors for a major insect group.
Collapse
Affiliation(s)
- Kevin J. Vogel
- Department of Entomology, The University of Georgia, Athens, GA, USA
- *Correspondence: Kevin J. Vogel, Department of Entomology, The University of Georgia, 413 Biological Sciences Building, Athens, GA 30602, USA e-mail:
| | - Mark R. Brown
- Department of Entomology, The University of Georgia, Athens, GA, USA
| | - Michael R. Strand
- Department of Entomology, The University of Georgia, Athens, GA, USA
| |
Collapse
|
21
|
Bonomi L, Brown M, Ungerleider N, Muse M, Matzuk MM, Schneyer A. Activin B regulates islet composition and islet mass but not whole body glucose homeostasis or insulin sensitivity. Am J Physiol Endocrinol Metab 2012; 303:E587-96. [PMID: 22739106 PMCID: PMC3468507 DOI: 10.1152/ajpendo.00177.2012] [Citation(s) in RCA: 17] [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] [Indexed: 01/18/2023]
Abstract
Based on the phenotype of the activin-like kinase-7 (ALK7)-null mouse, activins A and B have been proposed to play distinct roles in regulating pancreatic islet function and glucose homeostasis, with activin A acting to enhance islet function and insulin release while activin B antagonizes these actions. We therefore hypothesized that islets from activin B-null (BBKO) mice would have enhanced glucose-stimulated insulin secretion. In addition, we hypothesized that this enhanced islet function would translate into increased whole body glucose tolerance. We tested these hypotheses by analyzing glucose homeostasis, insulin secretion, and islet function in BBKO mice. No differences were observed in fasting glucose or insulin levels, glucose tolerance, or insulin sensitivity compared with weight-matched young or older males. Similarly, there were no significant differences in insulin secretion comparing islets from WT or BBKO males at either age. However, BBKO islets were more sensitive to activin A, myostatin (MSTN), and follistatin (FST) treatments, so that activin A and FST inhibited and MSTN enhanced glucose stimulated insulin secretion. While mean islet area and the distribution of islet areas were not different between the genotypes, islet mass, islet number, and the proportion of α-cells/islet were significantly reduced in BBKO islets. These results indicate that activin B does not antagonize activin A to influence whole body glucose homeostasis or β-cell function but does influence islet mass and proportion of α-cells/islet. Therefore, loss of activin B signaling alone does not account for the ALK7-null phenotype, but activin B may have important roles in modulating islet mass, islet number, and the cellular composition of islets.
Collapse
Affiliation(s)
- Lara Bonomi
- Pioneer Valley Life Science Institute, University of Massachusetts-Amherst, Springfield MA, USA
| | | | | | | | | | | |
Collapse
|
22
|
Wiater E, Vale W. Roles of activin family in pancreatic development and homeostasis. Mol Cell Endocrinol 2012; 359:23-9. [PMID: 22406274 DOI: 10.1016/j.mce.2012.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 02/14/2012] [Accepted: 02/15/2012] [Indexed: 01/15/2023]
Abstract
The transforming growth factor-beta (TGF-β) superfamily of ligands have been recognized as important signals in vertebrate embryonic development from the blastula stage to adulthood. In addition to roles in early development, TGF-β superfamily ligands, and particularly activin family ligands, are involved in specification, differentiation, and proliferation of multiple organ systems, including the pancreas. More recently, research has suggested that activin family ligands, binding proteins, receptors, and Smad signal transducers and modulators are involved in regulating adult pancreatic function and maintaining pancreatic islet homeostasis in the adult. This article will focus on outlining common themes in activin family regulation of embryonic pancreatic development and adult pancreatic homeostasis, particularly in activin family involvement in setting and maintaining populations of islet cells such as β-cells.
Collapse
Affiliation(s)
- Ezra Wiater
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute of Biological Studies, La Jolla, CA 92037, USA.
| | | |
Collapse
|