1
|
Phadnis A, Chawla D, Alex J, Jha P. Decoding MODY: exploring genetic roots and clinical pathways. Diabetol Int 2025; 16:257-271. [PMID: 40166432 PMCID: PMC11954780 DOI: 10.1007/s13340-025-00809-x] [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: 12/12/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025]
Abstract
Purpose Maturity-onset diabetes of the young (MODY) is a transformative factor in today's pattern of diabetes care. The definition of its genetic basis brings insight into the diabetes processes, opening up possibilities for its early detection through public health strategies and improvement in precision medicine. Current knowledge on MODY has been brought together in this review. Methods Extensive literature review on PubMed and Google Scholar databases was conducted. Studies encompassing (1) genetic underpinnings and their types, (2) the significance of its biomarkers, and (3) diagnostic techniques and treatment modalities were focused upon. Results The disease accounts for 1-2% of all cases of diabetes and is usually misdiagnosed as either Type 1 or Type 2 diabetes. Several genes are involved in the appropriate functioning of pancreatic β-cells and mutations in these genes lead to an impairment in glucose metabolism and insulin secretion. A mild degree of hyperglycaemia, but without ketosis, is typical of MODY, seen mostly in adolescents and young adults. Treatment varies, including sulfonylureas for HNF1A and HNF4A mutations, lifestyle management for GCK mutations, and emerging therapies like GLP1 receptor agonists. Conclusion Proper genetic diagnosis is cardinal to the best management of MODY. Genetic and clinical advances have been impressive in monogenic diabetes, but further research in novel therapies is needed to optimise outcomes with precision medicine.
Collapse
Affiliation(s)
- Anshuman Phadnis
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS Deemed to Be University, Mumbai, Maharashtra India
| | - Diya Chawla
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS Deemed to Be University, Mumbai, Maharashtra India
| | - Joanne Alex
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS Deemed to Be University, Mumbai, Maharashtra India
| | - Pamela Jha
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS Deemed to Be University, Mumbai, Maharashtra India
| |
Collapse
|
2
|
Wong A, Alejandro EU. Post translational modification regulation of transcription factors governing pancreatic β-cell identity and functional mass. Front Endocrinol (Lausanne) 2025; 16:1562646. [PMID: 40134803 PMCID: PMC11932907 DOI: 10.3389/fendo.2025.1562646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Dysfunction of the insulin-secreting β-cells is a key hallmark of Type 2 diabetes (T2D). In the natural history of the progression of T2D, factors such as genetics, early life exposures, lifestyle, and obesity dictate an individual's susceptibility risk to disease. Obesity is associated with insulin resistance and increased demand for insulin to maintain glucose homeostasis. Studies in both mouse and human islets have implicated the β-cell's ability to compensate through proliferation and survival (increasing functional β-cell mass) as a tipping point toward the development of disease. A growing body of evidence suggests the reduction of β-cell mass in T2D is driven majorly by loss of β-cell identity, rather than by apoptosis alone. The development and maintenance of pancreatic β-cell identity, function, and adaptation to stress is governed, in part, by the spatiotemporal expression of transcription factors (TFs), whose activity is regulated by signal-dependent post-translational modifications (PTM). In this review, we examine the role of these TFs in the developing pancreas and in the mature β-cell. We discuss functional implications of post-translational modifications on these transcription factors' activities and how an understanding of the pathways they regulate can inform therapies to promoteβ-cell regeneration, proliferation, and survival in diabetes.
Collapse
Affiliation(s)
- Alicia Wong
- Department of Genetics, Cell Biology, and Development, University of Minnesota Twin Cities, Minneapolis, MN, United States
| | - Emilyn U. Alejandro
- Department of Integrative Biology and Physiology, University of Minnesota Twin Cities, Minneapolis, MN, United States
| |
Collapse
|
3
|
Gilda JE, Nahar A, Kasiviswanathan D, Tropp N, Gilinski T, Lahav T, Alexandrovich D, Mandel-Gutfreund Y, Park S, Shemer S. Proteasome gene expression is controlled by coordinated functions of multiple transcription factors. J Cell Biol 2024; 223:e202402046. [PMID: 38767572 PMCID: PMC11104393 DOI: 10.1083/jcb.202402046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/14/2024] [Accepted: 04/16/2024] [Indexed: 05/22/2024] Open
Abstract
Proteasome activity is crucial for cellular integrity, but how tissues adjust proteasome content in response to catabolic stimuli is uncertain. Here, we demonstrate that transcriptional coordination by multiple transcription factors is required to increase proteasome content and activate proteolysis in catabolic states. Using denervated mouse muscle as a model system for accelerated proteolysis in vivo, we reveal that a two-phase transcriptional program activates genes encoding proteasome subunits and assembly chaperones to boost an increase in proteasome content. Initially, gene induction is necessary to maintain basal proteasome levels, and in a more delayed phase (7-10 days after denervation), it stimulates proteasome assembly to meet cellular demand for excessive proteolysis. Intriguingly, the transcription factors PAX4 and α-PALNRF-1 control the expression of proteasome among other genes in a combinatorial manner, driving cellular adaptation to muscle denervation. Consequently, PAX4 and α-PALNRF-1 represent new therapeutic targets to inhibit proteolysis in catabolic diseases (e.g., type-2 diabetes, cancer).
Collapse
Affiliation(s)
- Jennifer E Gilda
- Faculty of Biology, Technion Institute of Technology, Haifa, Israel
| | | | | | - Nadav Tropp
- Faculty of Biology, Technion Institute of Technology, Haifa, Israel
| | - Tamar Gilinski
- Faculty of Biology, Technion Institute of Technology, Haifa, Israel
| | - Tamar Lahav
- Faculty of Biology, Technion Institute of Technology, Haifa, Israel
| | | | | | | | - Shenhav Shemer
- Faculty of Biology, Technion Institute of Technology, Haifa, Israel
| |
Collapse
|
4
|
Narayan G, Ronima K R, Agrawal A, Thummer RP. An Insight into Vital Genes Responsible for β-cell Formation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1450:1-27. [PMID: 37432546 DOI: 10.1007/5584_2023_778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
The regulation of glucose homeostasis and insulin secretion by pancreatic β-cells, when disturbed, will result in diabetes mellitus. Replacement of dysfunctional or lost β-cells with fully functional ones can tackle the problem of β-cell generation in diabetes mellitus. Various pancreatic-specific genes are expressed during different stages of development, which have essential roles in pancreatogenesis and β-cell formation. These factors play a critical role in cellular-based studies like transdifferentiation or de-differentiation of somatic cells to multipotent or pluripotent stem cells and their differentiation into functional β-cells. This work gives an overview of crucial transcription factors expressed during various stages of pancreas development and their role in β-cell specification. In addition, it also provides a perspective on the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Gloria Narayan
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Ronima K R
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Akriti Agrawal
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.
| |
Collapse
|
5
|
Lau HH, Krentz NAJ, Abaitua F, Perez-Alcantara M, Chan JW, Ajeian J, Ghosh S, Lee Y, Yang J, Thaman S, Champon B, Sun H, Jha A, Hoon S, Tan NS, Gardner DSL, Kao SL, Tai ES, Gloyn AL, Teo AKK. PAX4 loss of function increases diabetes risk by altering human pancreatic endocrine cell development. Nat Commun 2023; 14:6119. [PMID: 37777536 PMCID: PMC10542369 DOI: 10.1038/s41467-023-41860-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 09/20/2023] [Indexed: 10/02/2023] Open
Abstract
The coding variant (p.Arg192His) in the transcription factor PAX4 is associated with an altered risk for type 2 diabetes (T2D) in East Asian populations. In mice, Pax4 is essential for beta cell formation but its role on human beta cell development and/or function is unknown. Participants carrying the PAX4 p.His192 allele exhibited decreased pancreatic beta cell function compared to homozygotes for the p.192Arg allele in a cross-sectional study in which we carried out an intravenous glucose tolerance test and an oral glucose tolerance test. In a pedigree of a patient with young onset diabetes, several members carry a newly identified p.Tyr186X allele. In the human beta cell model, EndoC-βH1, PAX4 knockdown led to impaired insulin secretion, reduced total insulin content, and altered hormone gene expression. Deletion of PAX4 in human induced pluripotent stem cell (hiPSC)-derived islet-like cells resulted in derepression of alpha cell gene expression. In vitro differentiation of hiPSCs carrying PAX4 p.His192 and p.X186 risk alleles exhibited increased polyhormonal endocrine cell formation and reduced insulin content that can be reversed with gene correction. Together, we demonstrate the role of PAX4 in human endocrine cell development, beta cell function, and its contribution to T2D-risk.
Collapse
Affiliation(s)
- Hwee Hui Lau
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Proteos, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Nicole A J Krentz
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Fernando Abaitua
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | - Jun-Wei Chan
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Proteos, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Jila Ajeian
- Oxford Centre for Diabetes Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Soumita Ghosh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Yunkyeong Lee
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jing Yang
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Swaraj Thaman
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Benoite Champon
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Han Sun
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alokkumar Jha
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Shawn Hoon
- Molecular Engineering Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Proteos, Singapore
| | - Nguan Soon Tan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | | | - Shih Ling Kao
- Department of Medicine, National University Hospital and National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - E Shyong Tai
- Department of Medicine, National University Hospital and National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Anna L Gloyn
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA.
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Proteos, Singapore.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
6
|
Ko J, Fonseca VA, Wu H. Pax4 in Health and Diabetes. Int J Mol Sci 2023; 24:8283. [PMID: 37175989 PMCID: PMC10179455 DOI: 10.3390/ijms24098283] [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: 04/03/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Paired box 4 (Pax4) is a key transcription factor involved in the embryonic development of the pancreatic islets of Langerhans. Consisting of a conserved paired box domain and a homeodomain, this transcription factor plays an essential role in early endocrine progenitor cells, where it is necessary for cell-fate commitment towards the insulin-secreting β cell lineage. Knockout of Pax4 in animal models leads to the absence of β cells, which is accompanied by a significant increase in glucagon-producing α cells, and typically results in lethality within days after birth. Mutations in Pax4 that cause an impaired Pax4 function are associated with diabetes pathogenesis in humans. In adulthood, Pax4 expression is limited to a distinct subset of β cells that possess the ability to proliferate in response to heightened metabolic needs. Upregulation of Pax4 expression is known to promote β cell survival and proliferation. Additionally, ectopic expression of Pax4 in pancreatic islet α cells or δ cells has been found to generate functional β-like cells that can improve blood glucose regulation in experimental diabetes models. Therefore, Pax4 represents a promising therapeutic target for the protection and regeneration of β cells in the treatment of diabetes. The purpose of this review is to provide a thorough and up-to-date overview of the role of Pax4 in pancreatic β cells and its potential as a therapeutic target for diabetes.
Collapse
Affiliation(s)
| | | | - Hongju Wu
- Section of Endocrinology, Department of Medicine, Tulane University Health Science Center, New Orleans, LA 70112, USA; (J.K.); (V.A.F.)
| |
Collapse
|
7
|
Gilda JE, Nahar A, Kasiviswanathan D, Tropp N, Gilinski T, Lahav T, Mandel-Gutfreund Y, Park S, Cohen S. Proteasome gene expression is controlled by the coordinated functions of multiple transcription factors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.12.536627. [PMID: 37205440 PMCID: PMC10187252 DOI: 10.1101/2023.04.12.536627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Proteasome activity is crucial for cellular integrity, but how tissues adjust proteasome content in response to catabolic stimuli is uncertain. Here, we demonstrate that transcriptional coordination by multiple transcription factors is required to increase proteasome content and activate proteolysis in catabolic states. Using denervated mouse muscle as a model system for accelerated proteolysis in vivo , we reveal that a two-phase transcriptional program activates genes encoding proteasome subunits and assembly chaperones to boost an increase in proteasome content. Initially, gene induction is necessary to maintain basal proteasome levels, and in a more delayed phase (7-10 d after denervation) it stimulates proteasome assembly to meet cellular demand for excessive proteolysis. Intriguingly, the transcription factors PAX4 and α-PAL NRF-1 control the expression of proteasome among other genes in a combinatorial manner, driving cellular adaptation to muscle denervation. Consequently, PAX4 and α-PAL NRF-1 represent new therapeutic targets to inhibit proteolysis in catabolic diseases (e.g. type-2 diabetes, cancer).
Collapse
|
8
|
Silva IBB, Kimura CH, Colantoni VP, Sogayar MC. Stem cells differentiation into insulin-producing cells (IPCs): recent advances and current challenges. Stem Cell Res Ther 2022; 13:309. [PMID: 35840987 PMCID: PMC9284809 DOI: 10.1186/s13287-022-02977-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 06/19/2022] [Indexed: 11/10/2022] Open
Abstract
Type 1 diabetes mellitus (T1D) is a chronic disease characterized by an autoimmune destruction of insulin-producing β-pancreatic cells. Although many advances have been achieved in T1D treatment, current therapy strategies are often unable to maintain perfect control of glycemic levels. Several studies are searching for new and improved methodologies for expansion of β-cell cultures in vitro to increase the supply of these cells for pancreatic islets replacement therapy. A promising approach consists of differentiation of stem cells into insulin-producing cells (IPCs) in sufficient number and functional status to be transplanted. Differentiation protocols have been designed using consecutive cytokines or signaling modulator treatments, at specific dosages, to activate or inhibit the main signaling pathways that control the differentiation of induced pluripotent stem cells (iPSCs) into pancreatic β-cells. Here, we provide an overview of the current approaches and achievements in obtaining stem cell-derived β-cells and the numerous challenges, which still need to be overcome to achieve this goal. Clinical translation of stem cells-derived β-cells for efficient maintenance of long-term euglycemia remains a major issue. Therefore, research efforts have been directed to the final steps of in vitro differentiation, aiming at production of functional and mature β-cells and integration of interdisciplinary fields to generate efficient cell therapy strategies capable of reversing the clinical outcome of T1D.
Collapse
Affiliation(s)
- Isaura Beatriz Borges Silva
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo, SP, 05360-130, Brazil.,Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Camila Harumi Kimura
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo, SP, 05360-130, Brazil
| | - Vitor Prado Colantoni
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo, SP, 05360-130, Brazil.,Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Mari Cleide Sogayar
- Cell and Molecular Therapy Center (NUCEL), School of Medicine, University of São Paulo, São Paulo, SP, 05360-130, Brazil. .,Department of Biochemistry, Chemistry Institute, University of São Paulo, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
9
|
Molecular Mechanism of Pancreatic β-Cell Failure in Type 2 Diabetes Mellitus. Biomedicines 2022; 10:biomedicines10040818. [PMID: 35453568 PMCID: PMC9030375 DOI: 10.3390/biomedicines10040818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 02/08/2023] Open
Abstract
Various important transcription factors in the pancreas are involved in the process of pancreas development, the differentiation of endocrine progenitor cells into mature insulin-producing pancreatic β-cells and the preservation of mature β-cell function. However, when β-cells are continuously exposed to a high glucose concentration for a long period of time, the expression levels of several insulin gene transcription factors are substantially suppressed, which finally leads to pancreatic β-cell failure found in type 2 diabetes mellitus. Here we show the possible underlying pathway for β-cell failure. It is likely that reduced expression levels of MafA and PDX-1 and/or incretin receptor in β-cells are closely associated with β-cell failure in type 2 diabetes mellitus. Additionally, since incretin receptor expression is reduced in the advanced stage of diabetes mellitus, incretin-based medicines show more favorable effects against β-cell failure, especially in the early stage of diabetes mellitus compared to the advanced stage. On the other hand, many subjects have recently suffered from life-threatening coronavirus infection, and coronavirus infection has brought about a new and persistent pandemic. Additionally, the spread of coronavirus infection has led to various limitations on the activities of daily life and has restricted economic development worldwide. It has been reported recently that SARS-CoV-2 directly infects β-cells through neuropilin-1, leading to apoptotic β-cell death and a reduction in insulin secretion. In this review article, we feature a possible molecular mechanism for pancreatic β-cell failure, which is often observed in type 2 diabetes mellitus. Finally, we are hopeful that coronavirus infection will decline and normal daily life will soon resume all over the world.
Collapse
|
10
|
Iida R, Ueki M, Yasuda T. Deficiency of M-LP/Mpv17L leads to development of β-cell hyperplasia and improved glucose tolerance via activation of the Wnt and TGF-β pathways. Biochim Biophys Acta Mol Basis Dis 2021; 1868:166318. [PMID: 34883249 DOI: 10.1016/j.bbadis.2021.166318] [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: 07/03/2021] [Revised: 11/17/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022]
Abstract
M-LP/Mpv17L is a protein that was initially identified during screening of age-dependently expressed genes in mice. We have recently demonstrated that M-LP/Mpv17L-knockout (M-LP/Mpv17L-KO) in human hepatoma cells leads to a reduction of cellular cyclic nucleotide phosphodiesterase (PDE) activity, and that in vitro-synthesized M-LP/Mpv17L possesses PDE activity. These findings suggest that M-LP/Mpv17L functions as an atypical PDE, even though it has none of the well-conserved catalytic region or other structural motifs characteristic of the PDE family. In this study, we found that M-LP/Mpv17L-KO mice developed β-cell hyperplasia and improved glucose tolerance. Deficiency of M-LP/Mpv17L in islets from KO mice at early postnatal stages or siRNA-mediated suppression of M-LP/Mpv17L in rat insulinoma cells led to marked upregulation of lymphoid enhancer binding factor 1 (Lef1) and transcription factor 7 (Tcf7), key nuclear effectors in the Wnt signaling pathway, and some of the factors essential for the development and maintenance of β-cells. Moreover, at the protein level, increases in the levels of phosphorylated β-catenin and glycogen synthase kinase-3β (GSK-3β) were observed, indicating activation of the Wnt and TGF-β signaling pathways. Taken together, these findings suggest that protein kinase A (PKA)-dependent phosphorylations of β-catenin and GSK-3β, the key mediators of the Wnt and/or TGF-β signaling pathways, are the most upstream events triggering β-cell hyperplasia and improved glucose tolerance caused by M-LP/Mpv17L deficiency.
Collapse
Affiliation(s)
- Reiko Iida
- Life Science Unit, School of Medical Sciences, University of Fukui, Fukui 910-1193, Japan; Life Science Innovation Center, University of Fukui, Fukui 910-1193, Japan.
| | - Misuzu Ueki
- Molecular Neuroscience Unit, School of Medical Sciences, University of Fukui, Fukui 910-1193, Japan
| | - Toshihiro Yasuda
- Life Science Innovation Center, University of Fukui, Fukui 910-1193, Japan
| |
Collapse
|
11
|
Yong J, Johnson JD, Arvan P, Han J, Kaufman RJ. Therapeutic opportunities for pancreatic β-cell ER stress in diabetes mellitus. Nat Rev Endocrinol 2021; 17:455-467. [PMID: 34163039 PMCID: PMC8765009 DOI: 10.1038/s41574-021-00510-4] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
Diabetes mellitus is characterized by the failure of insulin-secreting pancreatic β-cells (or β-cell death) due to either autoimmunity (type 1 diabetes mellitus) or failure to compensate for insulin resistance (type 2 diabetes mellitus; T2DM). In addition, mutations of critical genes cause monogenic diabetes. The endoplasmic reticulum (ER) is the primary site for proinsulin folding; therefore, ER proteostasis is crucial for both β-cell function and survival under physiological and pathophysiological challenges. Importantly, the ER is also the major intracellular Ca2+ storage organelle, generating Ca2+ signals that contribute to insulin secretion. ER stress is associated with the pathogenesis of diabetes mellitus. In this Review, we summarize the mutations in monogenic diabetes that play causal roles in promoting ER stress in β-cells. Furthermore, we discuss the possible mechanisms responsible for ER proteostasis imbalance with a focus on T2DM, in which both genetics and environment are considered important in promoting ER stress in β-cells. We also suggest that controlled insulin secretion from β-cells might reduce the progression of a key aspect of the metabolic syndrome, namely nonalcoholic fatty liver disease. Finally, we evaluate potential therapeutic approaches to treat T2DM, including the optimization and protection of functional β-cell mass in individuals with T2DM.
Collapse
Affiliation(s)
- Jing Yong
- Degenerative Diseases Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - James D Johnson
- Department of Cellular and Physiological Sciences & Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Peter Arvan
- Division of Metabolism Endocrinology & Diabetes, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jaeseok Han
- Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan-si, Choongchungnam-do, Republic of Korea.
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
12
|
Demirci DK, Darendeliler F, Poyrazoglu S, Al ADK, Gul N, Tutuncu Y, Gulfidan G, Arga KY, Cacina C, Ozturk O, Aydogan HY, Satman I. Monogenic Childhood Diabetes: Dissecting Clinical Heterogeneity by Next-Generation Sequencing in Maturity-Onset Diabetes of the Young. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:431-449. [PMID: 34171966 DOI: 10.1089/omi.2021.0081] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Diabetes is a common disorder with a heterogeneous clinical presentation and an enormous burden on health care worldwide. About 1-6% of patients with diabetes suffer from maturity-onset diabetes of the young (MODY), the most common form of monogenic diabetes with autosomal dominant inheritance. MODY is genetically and clinically heterogeneous and caused by genetic variations in pancreatic β-cell development and insulin secretion. We report here new findings from targeted next-generation sequencing (NGS) of 13 MODY-related genes. A sample of 22 unrelated pediatric patients with MODY and 13 unrelated healthy controls were recruited from a Turkish population. Targeted NGS was performed with Miseq 4000 (Illumina) to identify genetic variations in 13 MODY-related genes: HNF4A, GCK, HNF1A, PDX1, HNF1B, NEUROD1, KLF11, CEL, PAX4, INS, BLK, ABCC8, and KCNJ11. The NGS data were analyzed adhering to the Genome Analysis ToolKit (GATK) best practices pipeline, and variant filtering and annotation were performed. In the patient sample, we identified 43 MODY-specific genetic variations that were not present in the control group, including 11 missense mutations and 4 synonymous mutations. Importantly, and to the best of our knowledge, the missense mutations NEUROD1 p.D202E, KFL11 p.R461Q, BLK p.G248R, and KCNJ11 p.S385F were first associated with MODY in the present study. These findings contribute to the worldwide knowledge base on MODY and molecular correlates of clinical heterogeneity in monogenic childhood diabetes. Further comparative population genetics and functional genomics studies are called for, with an eye to discovery of novel diagnostics and personalized medicine in MODY. Because MODY is often misdiagnosed as type 1 or type 2 diabetes mellitus, advances in MODY diagnostics with NGS stand to benefit diabetes overall clinical care as well.
Collapse
Affiliation(s)
- Deniz Kanca Demirci
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Halic University, Istanbul, Turkey.,Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Feyza Darendeliler
- Pediatric Endocrinology Unit, Department of Pediatrics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sukran Poyrazoglu
- Pediatric Endocrinology Unit, Department of Pediatrics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Asli Derya Kardelen Al
- Pediatric Endocrinology Unit, Department of Pediatrics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Nurdan Gul
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Yildiz Tutuncu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.,Department of Immunology, School of Medicine, KUTTAM, Koc University, Istanbul, Turkey
| | - Gizem Gulfidan
- Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul, Turkey
| | - Kazim Yalcin Arga
- Department of Bioengineering, Faculty of Engineering, Marmara University, Istanbul, Turkey.,Institute of Public Health and Chronic Diseases, The Health Institutes of Turkey, Istanbul, Turkey
| | - Canan Cacina
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Oguz Ozturk
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Hulya Yilmaz Aydogan
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Ilhan Satman
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.,Institute of Public Health and Chronic Diseases, The Health Institutes of Turkey, Istanbul, Turkey
| |
Collapse
|
13
|
Sanchez Caballero L, Gorgogietas V, Arroyo MN, Igoillo-Esteve M. Molecular mechanisms of β-cell dysfunction and death in monogenic forms of diabetes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 359:139-256. [PMID: 33832649 DOI: 10.1016/bs.ircmb.2021.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Monogenetic forms of diabetes represent 1%-5% of all diabetes cases and are caused by mutations in a single gene. These mutations, that affect genes involved in pancreatic β-cell development, function and survival, or insulin regulation, may be dominant or recessive, inherited or de novo. Most patients with monogenic diabetes are very commonly misdiagnosed as having type 1 or type 2 diabetes. The severity of their symptoms depends on the nature of the mutation, the function of the affected gene and, in some cases, the influence of additional genetic or environmental factors that modulate severity and penetrance. In some patients, diabetes is accompanied by other syndromic features such as deafness, blindness, microcephaly, liver and intestinal defects, among others. The age of diabetes onset may also vary from neonatal until early adulthood manifestations. Since the different mutations result in diverse clinical presentations, patients usually need different treatments that range from just diet and exercise, to the requirement of exogenous insulin or other hypoglycemic drugs, e.g., sulfonylureas or glucagon-like peptide 1 analogs to control their glycemia. As a consequence, awareness and correct diagnosis are crucial for the proper management and treatment of monogenic diabetes patients. In this chapter, we describe mutations causing different monogenic forms of diabetes associated with inadequate pancreas development or impaired β-cell function and survival, and discuss the molecular mechanisms involved in β-cell demise.
Collapse
Affiliation(s)
- Laura Sanchez Caballero
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Vyron Gorgogietas
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Maria Nicol Arroyo
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/
| | - Mariana Igoillo-Esteve
- ULB Center for Diabetes Research (UCDR), Université Libre de Bruxelles, Brussels, Belgium. http://www.ucdr.be/.
| |
Collapse
|
14
|
Gao A, Gu B, Zhang J, Fang C, Su J, Li H, Han R, Ye L, Wang W, Ning G, Wang J, Gu W. Missense Variants in PAX4 Are Associated with Early-Onset Diabetes in Chinese. Diabetes Ther 2021; 12:289-300. [PMID: 33216280 PMCID: PMC7843779 DOI: 10.1007/s13300-020-00960-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 10/27/2020] [Indexed: 11/05/2022] Open
Abstract
INTRODUCTION East Asians are more susceptible to early-onset diabetes than Europeans and exhibit reduced insulin secretion at earlier stages. PAX4 plays a critical role in the development of β-cells. The dysfunction-missense variants PAX4 R192H and PAX4 R192S are common in East Asians but rare in Europeans. Therefore, we aim to investigate the diabetes-associated genes, including PAX4 R192H/S, in East Asians with early-onset diabetes. METHODS Exome variants of 80 Chinese early-onset diabetes patients (onset age < 35 years) after the exclusion of type 1 diabetes (T1D) were detected by a customized gene panel covering 32 known diabetes-associated genes. Then, 229 subjects with early-onset diabetes (T1D excluded) and 1679 controls from the Chinese population were genotyped to validate the association of PAX4 R192H/S with early-onset diabetes and related phenotypes. RESULTS The gene panel detected 11 monogenic diabetes patients with five novel mutations among the 80 early-onset diabetes patients. Asian-specifically enriched PAX4 R192H and R192S were associated with early-onset diabetes (R192H: OR 1.88, 95% CI 1.37-2.60, P = 8.41 × 10-5; R192S: OR 1.71, 95% CI 1.17-2.51, P = 0.005). In early-onset diabetes patients, PAX4 R192H carriers had higher haemoglobin A1c (HbA1c) levels (P = 0.030) and lower 2 h C-peptide levels in the oral glucose tolerance test (OGTT) (P = 0.040); R192S carriers had lower fasting C-peptide (FCP) (P = 0.011) and 2 h C-peptide levels (P = 0.033) in OGTT than non-variant carriers. CONCLUSIONS The ethnic-specific enrichment of PAX4 R192H/S predisposing East Asians to early-onset diabetes with decreased C-peptide levels may be one explanation of the discrepancy of diabetes between East Asians and Europeans. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov (NCT01938365).
Collapse
Affiliation(s)
- Aibo Gao
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200025, China
- National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Bin Gu
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200025, China
| | - Juan Zhang
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200025, China
| | - Chen Fang
- The Second Affiliated Hospital of Soochow University, Soochow, 215004, China
| | - Junlei Su
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200025, China
| | - Haorong Li
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200025, China
| | - Rulai Han
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200025, China
| | - Lei Ye
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200025, China
| | - Weiqing Wang
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200025, China
| | - Guang Ning
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200025, China
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200025, China.
| | - Weiqiong Gu
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, 200025, China.
| |
Collapse
|
15
|
The epidemiology, molecular pathogenesis, diagnosis, and treatment of maturity-onset diabetes of the young (MODY). Clin Diabetes Endocrinol 2020; 6:20. [PMID: 33292863 PMCID: PMC7640483 DOI: 10.1186/s40842-020-00112-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
Background The most common type of monogenic diabetes is maturity-onset diabetes of the young (MODY), a clinically and genetically heterogeneous group of endocrine disorders that affect 1–5% of all patients with diabetes mellitus. MODY is characterized by autosomal dominant inheritance but de novo mutations have been reported. Clinical features of MODY include young-onset hyperglycemia, evidence of residual pancreatic function, and lack of beta cell autoimmunity or insulin resistance. Glucose-lowering medications are the main treatment options for MODY. The growing recognition of the clinical and public health significance of MODY by clinicians, researchers, and governments may lead to improved screening and diagnostic practices. Consequently, this review article aims to discuss the epidemiology, pathogenesis, diagnosis, and treatment of MODY based on relevant literature published from 1975 to 2020. Main body The estimated prevalence of MODY from European cohorts is 1 per 10,000 in adults and 1 per 23,000 in children. Since little is known about the prevalence of MODY in African, Asian, South American, and Middle Eastern populations, further research in non-European cohorts is needed to help elucidate MODY’s exact prevalence. Currently, 14 distinct subtypes of MODY can be diagnosed through clinical assessment and genetic analysis. Various genetic mutations and disease mechanisms contribute to the pathogenesis of MODY. Management of MODY is subtype-specific and includes diet, oral antidiabetic drugs, or insulin. Conclusions Incidence and prevalence estimates for MODY are derived from epidemiologic studies of young people with diabetes who live in Europe, Australia, and North America. Mechanisms involved in the pathogenesis of MODY include defective transcriptional regulation, abnormal metabolic enzymes, protein misfolding, dysfunctional ion channels, or impaired signal transduction. Clinicians should understand the epidemiology and pathogenesis of MODY because such knowledge is crucial for accurate diagnosis, individualized patient management, and screening of family members.
Collapse
|
16
|
Melnikova AI, Krasnova TS, Zubkova NA, Tiulpakov AN, Rubtsov PM. Alternative Variants of Pax4 Human Transcription Factor: Comparative Transcriptional Activity. Mol Biol 2020. [DOI: 10.1134/s0026893320050076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
17
|
Sussman W, Stevenson M, Mowdawalla C, Mota S, Ragolia L, Pan X. BMAL1 controls glucose uptake through paired-homeodomain transcription factor 4 in differentiated Caco-2 cells. Am J Physiol Cell Physiol 2019; 317:C492-C501. [PMID: 31216190 PMCID: PMC6766619 DOI: 10.1152/ajpcell.00058.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 02/08/2023]
Abstract
The transcription factor aryl hydrocarbon receptor nuclear translocator-like protein-1 (BMAL1) is an essential regulator of the circadian clock, which controls the 24-h cycle of physiological processes such as nutrient absorption. To examine the role of BMAL1 in small intestinal glucose absorption, we used differentiated human colon adenocarcinoma cells (Caco-2 cells). Here, we show that BMAL1 regulates glucose uptake in differentiated Caco-2 cells and that this process is dependent on the glucose transporter sodium-glucose cotransporter 1 (SGLT1). Mechanistic studies show that BMAL1 regulates glucose uptake by controlling the transcription of SGLT1 involving paired-homeodomain transcription factor 4 (PAX4), a transcriptional repressor. This is supported by the observation that clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated endonuclease Cas9 (Cas9) knockdown of PAX4 increases SGLT1 and glucose uptake. Chromatin immunoprecipitation (ChIP) and ChIP-quantitative PCR assays show that the knockdown or overexpression of BMAL1 decreases or increases the binding of PAX4 to the hepatocyte nuclear factor 1-α binding site of the SGLT1 promoter, respectively. These findings identify BMAL1 as a critical mediator of small intestine carbohydrate absorption and SGLT1.
Collapse
Affiliation(s)
- Whitney Sussman
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, New York
| | - Matthew Stevenson
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, New York
| | - Cyrus Mowdawalla
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, New York
| | - Samantha Mota
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, New York
| | - Louis Ragolia
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, New York
| | - Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, New York
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, New York
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, New York
| |
Collapse
|
18
|
Singh A, Gibert Y, Dwyer KM. The adenosine, adrenergic and opioid pathways in the regulation of insulin secretion, beta cell proliferation and regeneration. Pancreatology 2018; 18:615-623. [PMID: 29937364 DOI: 10.1016/j.pan.2018.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/25/2018] [Accepted: 06/19/2018] [Indexed: 02/07/2023]
Abstract
Insulin, a key hormone produced by pancreatic beta cells precisely regulates glucose metabolism in vertebrates. In type 1 diabetes, the beta cell mass is destroyed, a process triggered by a combination of environmental and genetic factors. This ultimately results in absolute insulin deficiency and dysregulated glucose metabolism resulting in a number of detrimental pathophysiological effects. The traditional focus of treating type 1 diabetes has been to control blood sugar levels through the administration of exogenous insulin. Newer approaches aim to replace the beta cell mass through pancreatic or islet transplantation. Type 2 diabetes results from a relative insulin deficiency for the prevailing insulin resistance. Treatments are generally aimed at reducing insulin resistance and/or augmenting insulin secretion and the use of insulin itself is often required. It is increasingly being recognized that the beta cell mass is dynamic and increases insulin secretion in response to beta cell mitogens and stress signals to maintain glycemia within a very narrow physiological range. This review critically discusses the role of adrenergic, adenosine and opioid pathways and their interrelationship in insulin secretion, beta cell proliferation and regeneration.
Collapse
Affiliation(s)
- Amitoj Singh
- Deakin University, School of Medicine, Faculty of Health, 75 Pigdons Rd, Waurn Ponds, Geelong, VIC, 3216, Australia
| | - Yann Gibert
- Deakin University, School of Medicine, Faculty of Health, 75 Pigdons Rd, Waurn Ponds, Geelong, VIC, 3216, Australia
| | - Karen M Dwyer
- Deakin University, School of Medicine, Faculty of Health, 75 Pigdons Rd, Waurn Ponds, Geelong, VIC, 3216, Australia.
| |
Collapse
|
19
|
Malenczyk K, Szodorai E, Schnell R, Lubec G, Szabó G, Hökfelt T, Harkany T. Secretagogin protects Pdx1 from proteasomal degradation to control a transcriptional program required for β cell specification. Mol Metab 2018; 14:108-120. [PMID: 29910119 PMCID: PMC6034064 DOI: 10.1016/j.molmet.2018.05.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Specification of endocrine cell lineages in the developing pancreas relies on extrinsic signals from non-pancreatic tissues, which initiate a cell-autonomous sequence of transcription factor activation and repression switches. The steps in this pathway share reliance on activity-dependent Ca2+ signals. However, the mechanisms by which phasic Ca2+ surges become converted into a dynamic, cell-state-specific and physiologically meaningful code made up by transcription factors constellations remain essentially unknown. METHODS We used high-resolution histochemistry to explore the coincident expression of secretagogin and transcription factors driving β cell differentiation. Secretagogin promoter activity was tested in response to genetically manipulating Pax6 and Pax4 expression. Secretagogin null mice were produced with their pancreatic islets morphologically and functionally characterized during fetal development. A proteomic approach was utilized to identify the Ca2+-dependent interaction of secretagogin with subunits of the 26S proteasome and verified in vitro by focusing on Pdx1 retention. RESULTS Here, we show that secretagogin, a Ca2+ sensor protein that controls α and β cell turnover in adult, is in fact expressed in endocrine pancreas from the inception of lineage segregation in a Pax4-and Pax6-dependent fashion. By genetically and pharmacologically manipulating secretagogin expression and interactome engagement in vitro, we find secretagogin to gate excitation-driven Ca2+ signals for β cell differentiation and insulin production. Accordingly, secretagogin-/- fetuses retain a non-committed pool of endocrine progenitors that co-express both insulin and glucagon. We identify the Ca2+-dependent interaction of secretagogin with subunits of the 26S proteasome complex to prevent Pdx1 degradation through proteasome inactivation. This coincides with retained Nkx6.1, Pax4 and insulin transcription in prospective β cells. CONCLUSIONS In sum, secretagogin scales the temporal availability of a Ca2+-dependent transcription factor network to define β cell identity.
Collapse
Affiliation(s)
- Katarzyna Malenczyk
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090, Vienna, Austria; Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-17177, Stockholm, Sweden
| | - Edit Szodorai
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090, Vienna, Austria; Paracelsus Medical University, Strubergasse 21, A-5020, Salzburg, Austria
| | - Robert Schnell
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, SE-17177, Stockholm, Sweden
| | - Gert Lubec
- Paracelsus Medical University, Strubergasse 21, A-5020, Salzburg, Austria
| | - Gábor Szabó
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony utca 43, H-1083, Budapest, Hungary
| | - Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-17177, Stockholm, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090, Vienna, Austria; Department of Neuroscience, Karolinska Institutet, Retzius väg 8, SE-17177, Stockholm, Sweden.
| |
Collapse
|
20
|
Targeted Mutation of NGN3 Gene Disrupts Pancreatic Endocrine Cell Development in Pigs. Sci Rep 2018; 8:3582. [PMID: 29483633 PMCID: PMC5827570 DOI: 10.1038/s41598-018-22050-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/15/2018] [Indexed: 12/12/2022] Open
Abstract
The domestic pig is an attractive model for biomedical research because of similarities in anatomy and physiology to humans. However, key gaps remain in our understanding of the role of developmental genes in pig, limiting its full potential. In this publication, the role of NEUROGENIN 3 (NGN3), a transcription factor involved in endocrine pancreas development has been investigated by CRISPR/Cas9 gene ablation. Precomplexed Cas9 ribonucleoproteins targeting NGN3 were injected into in vivo derived porcine embryos, and transferred into surrogate females. On day 60 of pregnancy, nine fetuses were collected for genotypic and phenotypic analysis. One of the piglets was identified as an in-frame biallelic knockout (Δ2/Δ2), which showed a loss of putative NGN3-downstream target genes: NEUROD1 and PAX4, as well as insulin, glucagon, somatostatin and pancreatic polypeptide-Y. Fibroblasts from this fetus were used in somatic cell nuclear transfer to generate clonal animals to qualify the effect of mutation on embryonic lethality. Three live piglets were born, received colostrum and suckled normally, but experienced extreme weight loss over a 24 to 36-hour period requiring humane euthanasia. Expression of pancreatic endocrine hormones: insulin, glucagon, and somatostatin were lost. The data support a critical role of NGN3 in porcine endocrine pancreas development.
Collapse
|
21
|
Firdous P, Nissar K, Ali S, Ganai BA, Shabir U, Hassan T, Masoodi SR. Genetic Testing of Maturity-Onset Diabetes of the Young Current Status and Future Perspectives. Front Endocrinol (Lausanne) 2018; 9:253. [PMID: 29867778 PMCID: PMC5966560 DOI: 10.3389/fendo.2018.00253] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 05/02/2018] [Indexed: 12/12/2022] Open
Abstract
Diabetes is a global epidemic problem growing exponentially in Asian countries posing a serious threat. Among diabetes, maturity-onset diabetes of the young (MODY) is a heterogeneous group of monogenic disorders that occurs due to β cell dysfunction. Genetic defects in the pancreatic β-cells result in the decrease of insulin production required for glucose utilization thereby lead to early-onset diabetes (often <25 years). It is generally considered as non-insulin dependent form of diabetes and comprises of 1-5% of total diabetes. Till date, 14 genes have been identified and mutation in them may lead to MODY. Different genetic testing methodologies like linkage analysis, restriction fragment length polymorphism, and DNA sequencing are used for the accurate and correct investigation of gene mutations associated with MODY. The next-generation sequencing has emerged as one of the most promising and effective tools to identify novel mutated genes related to MODY. Diagnosis of MODY is mainly relying on the sequential screening of the three marker genes like hepatocyte nuclear factor 1 alpha (HNF1α), hepatocyte nuclear factor 4 alpha (HNF4α), and glucokinase (GCK). Interestingly, MODY patients can be managed by diet alone for many years and may also require minimal doses of sulfonylureas. The primary objective of this article is to provide a review on current status of MODY, its prevalence, genetic testing/diagnosis, possible treatment, and future perspective.
Collapse
Affiliation(s)
- Parveena Firdous
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, India
| | - Kamran Nissar
- Department of Biochemistry, University of Kashmir, Srinagar, India
| | - Sajad Ali
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, India
| | - Bashir Ahmad Ganai
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, India
- *Correspondence: Bashir Ahmad Ganai,
| | - Uzma Shabir
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, India
| | - Toyeeba Hassan
- Centre of Research for Development (CORD), University of Kashmir, Srinagar, India
| | - Shariq Rashid Masoodi
- Department of Endocrinology, Sher-I-Kashmir Institute of Medical Sciences, Srinagar, India
| |
Collapse
|
22
|
Wong P, Markey M, Rapp CM, Darrow RM, Ziesel A, Organisciak D. Enhancing the efficacy of AREDS antioxidants in light-induced retinal degeneration. Mol Vis 2017; 23:718-739. [PMID: 29062223 PMCID: PMC5640517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 10/08/2017] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Light-induced photoreceptor cell degeneration and disease progression in age-related macular degeneration (AMD) involve oxidative stress and visual cell loss, which can be prevented, or slowed, by antioxidants. Our goal was to test the protective efficacy of a traditional Age-related Eye Disease Study antioxidant formulation (AREDS) and AREDS combined with non-traditional antioxidants in a preclinical animal model of photooxidative retinal damage. METHODS Male Sprague-Dawley rats were reared in a low-intensity (20 lux) or high-intensity (200 lux) cyclic light environment for 6 weeks. Some animals received a daily dietary supplement consisting of a small cracker infused with an AREDS antioxidant mineral mixture, AREDS antioxidants minus zinc, or zinc oxide alone. Other rats received AREDS combined with a detergent extract of the common herb rosemary, AREDS plus carnosic acid, zinc oxide plus rosemary, or rosemary alone. Antioxidant efficacy was determined by measuring retinal DNA levels 2 weeks after 6 h of intense exposure to white light (9,000 lux). Western blotting was used to determine visual cell opsin and arrestin levels following intense light treatment. Rhodopsin regeneration was determined after 1 h of exposure to light. Gene array analysis was used to determine changes in the expression of retinal genes resulting from light rearing environment or from antioxidant supplementation. RESULTS Chronic high-intensity cyclic light rearing resulted in lower levels of rod and cone opsins, retinal S-antigen (S-ag), and medium wavelength cone arrestin (mCAR) than found for rats maintained in low cyclic light. However, as determined by retinal DNA, and by residual opsin and arrestin levels, 2 weeks after acute photooxidative damage, visual cell loss was greater in rats reared in low cyclic light. Retinal damage decreased with AREDS plus rosemary, or with zinc oxide plus rosemary whereas AREDS alone and zinc oxide alone (at their daily recommended levels) were both ineffective. One week of supplemental AREDS plus carnosic acid resulted in higher levels of rod and cone cell proteins, and higher levels of retinal DNA than for AREDS alone. Rhodopsin regeneration was unaffected by the rosemary treatment. Retinal gene array analysis showed reduced expression of medium- wavelength opsin 1 and arrestin C in the high-light reared rats versus the low-light rats. The transition of rats from low cyclic light to a high cyclic light environment resulted in the differential expression of 280 gene markers, enriched for genes related to inflammation, apoptosis, cytokine, innate immune response, and receptors. Rosemary, zinc oxide plus rosemary, and AREDS plus rosemary suppressed 131, 241, and 266 of these genes (respectively) in high-light versus low-light animals and induced a small subset of changes in gene expression that were independent of light rearing conditions. CONCLUSIONS Long-term environmental light intensity is a major determinant of retinal gene and protein expression, and of visual cell survival following acute photooxidative insult. Rats preconditioned by high-light rearing exhibit lower levels of cone opsin mRNA and protein, and lower mCAR protein, than low-light reared animals, but greater retention of retinal DNA and proteins following photooxidative damage. Rosemary enhanced the protective efficacy of AREDS and led to the greatest effect on the retinal genome in animals reared in high environmental light. Chronic administration of rosemary antioxidants may be a useful adjunct to the therapeutic benefit of AREDS in slowing disease progression in AMD.
Collapse
Affiliation(s)
- Paul Wong
- Department of Ophthalmology, Emory University, Atlanta, GA
| | - M. Markey
- Center for Genomics Research; Wright State University, Dayton, OH
| | - C. M. Rapp
- Petticrew Research Laboratory, Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH
| | - R. M. Darrow
- Petticrew Research Laboratory, Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH
| | - A. Ziesel
- Department of Ophthalmology, Emory University, Atlanta, GA
| | - D.T. Organisciak
- Petticrew Research Laboratory, Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH
| |
Collapse
|
23
|
Krentz NAJ, van Hoof D, Li Z, Watanabe A, Tang M, Nian C, German MS, Lynn FC. Phosphorylation of NEUROG3 Links Endocrine Differentiation to the Cell Cycle in Pancreatic Progenitors. Dev Cell 2017; 41:129-142.e6. [PMID: 28441528 DOI: 10.1016/j.devcel.2017.02.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 10/28/2016] [Accepted: 02/09/2017] [Indexed: 02/08/2023]
Abstract
During pancreatic development, proliferating pancreatic progenitors activate the proendocrine transcription factor neurogenin 3 (NEUROG3), exit the cell cycle, and differentiate into islet cells. The mechanisms that direct robust NEUROG3 expression within a subset of progenitor cells control the size of the endocrine population. Here we demonstrate that NEUROG3 is phosphorylated within the nucleus on serine 183, which catalyzes its hyperphosphorylation and proteosomal degradation. During progression through the progenitor cell cycle, NEUROG3 phosphorylation is driven by the actions of cyclin-dependent kinases 2 and 4/6 at G1/S cell-cycle checkpoint. Using models of mouse and human pancreas development, we show that lengthening of the G1 phase of the pancreatic progenitor cell cycle is essential for proper induction of NEUROG3 and initiation of endocrine cell differentiation. In sum, these studies demonstrate that progenitor cell-cycle G1 lengthening, through its actions on stabilization of NEUROG3, is an essential variable in normal endocrine cell genesis.
Collapse
Affiliation(s)
- Nicole A J Krentz
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, 950 28th Avenue West, Vancouver, BC V5Z 4H4, Canada
| | - Dennis van Hoof
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research and Diabetes Center, University of California San Francisco, San Francisco, CA 94143-0669, USA
| | - Zhongmei Li
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research and Diabetes Center, University of California San Francisco, San Francisco, CA 94143-0669, USA
| | - Akie Watanabe
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, 950 28th Avenue West, Vancouver, BC V5Z 4H4, Canada
| | - Mei Tang
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, 950 28th Avenue West, Vancouver, BC V5Z 4H4, Canada
| | - Cuilan Nian
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, 950 28th Avenue West, Vancouver, BC V5Z 4H4, Canada
| | - Michael S German
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research and Diabetes Center, University of California San Francisco, San Francisco, CA 94143-0669, USA; Department of Medicine, University of California San Francisco, 35 Medical Center Way, RMB 1025, San Francisco, CA 94143-0669, USA.
| | - Francis C Lynn
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, 950 28th Avenue West, Vancouver, BC V5Z 4H4, Canada.
| |
Collapse
|
24
|
Al-Khawaga S, Memon B, Butler AE, Taheri S, Abou-Samra AB, Abdelalim EM. Pathways governing development of stem cell-derived pancreatic β cells: lessons from embryogenesis. Biol Rev Camb Philos Soc 2017. [DOI: 10.1111/brv.12349] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Sara Al-Khawaga
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| | - Bushra Memon
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| | - Alexandra E. Butler
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine; University of California; Los Angeles CA 90095 U.S.A
| | - Shahrad Taheri
- Department of Medicine; Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, PO BOX 24144; Doha Qatar
- Department of Medicine; Qatar Metabolic Institute, Hamad Medical Corporation; Doha Qatar
| | - Abdul B. Abou-Samra
- Department of Medicine; Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, PO BOX 24144; Doha Qatar
- Department of Medicine; Qatar Metabolic Institute, Hamad Medical Corporation; Doha Qatar
| | - Essam M. Abdelalim
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| |
Collapse
|
25
|
Abstract
A small number of cells in the adult pancreas are endocrine cells. They are arranged in clusters called islets of Langerhans. The islets make insulin, glucagon, and other endocrine hormones, and release them into the blood circulation. These hormones help control the level of blood glucose. Therefore, a dysfunction of endocrine cells in the pancreas results in impaired glucose homeostasis, or diabetes mellitus. The pancreas is an organ that originates from the evaginations of pancreatic progenitor cells in the epithelium of the foregut endoderm. Pancreas organogenesis and maturation of the islets of Langerhans occurs via a coordinated and complex interplay of transcriptional networks and signaling molecules, which guide a stepwise and repetitive process of the propagation of progenitor cells and their maturation, eventually resulting in a fully functional organ. Increasing our understanding of the extrinsic, as well as intrinsic mechanisms that control these processes should facilitate the efforts to generate surrogate β cells from ES or iPS cells, or to reactivate the function of important cell types within pancreatic islets that are lost in diabetes.
Collapse
Affiliation(s)
- Yoshio Fujitani
- Laboratory of Developmental Biology & Metabolism, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan
- AMED-CREST Program, Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan
- Department of Metabolism and Endocrinology, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| |
Collapse
|
26
|
Xu L, Xu C, Zhou S, Liu X, Wang J, Liu X, Qian S, Xin Y, Gao Y, Zhu Y, Tang X. PAX4 promotes PDX1-induced differentiation of mesenchymal stem cells into insulin-secreting cells. Am J Transl Res 2017; 9:874-886. [PMID: 28386318 PMCID: PMC5375983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 01/31/2017] [Indexed: 06/07/2023]
Abstract
A shortage of postmortem pancreatic tissue for islet isolation impedes the application of cell replacement therapy in patients with diabetes. As an alternative for islet cell transplantation, transcription factors, including PDX1, PAX4, and neurogenin-3, that aid in the formation of insulin-producing β cells during development have been investigated. The present study evaluated the effects of PAX4 and PDX1 on the differentiation of mesenchymal stem cells (MSCs) into insulin-producing β-like cells in vitro using recombinant adenoviruses carrying PDX1 or PDX1 plus PAX4. RT-PCR, Western blot, and immunofluorescence assays were used to detect the expression levels of relevant genes and proteins, and enzyme-linked immunosorbent assays were used to determine the amount of insulin and C-peptide secreted by the virus-infected cells following stimulation with high glucose. The results showed that PAX4 markedly enhanced the propensity of PDX1-positive MSCs to form mature islet-like clusters and functional insulin-producing β-like cells. Our findings provide a novel foundation for generating β-like cells from MSCs with PAX4 and PDX1 for future clinical application.
Collapse
Affiliation(s)
- Lifa Xu
- Stem Cell Engineering Research Center, School of Medical, Anhui University of Science & TechnologyHuainan 232001, P.R. China
| | - Congjing Xu
- Department of Respiration, Tumour Hospital of Affiliated Huainan Oriental Hospital Group, Anhui University of Science and TechnologyHuainan 232035, P.R. China
| | - Shuping Zhou
- Huainan First People’s Hospital and First Affiliated Hospital of Medical College, Anhui University of Science & TechnologyHuainan 232001, P.R. China
| | - Xueke Liu
- Stem Cell Engineering Research Center, School of Medical, Anhui University of Science & TechnologyHuainan 232001, P.R. China
| | - Jian Wang
- Stem Cell Engineering Research Center, School of Medical, Anhui University of Science & TechnologyHuainan 232001, P.R. China
| | - Xinkuang Liu
- Huainan First People’s Hospital and First Affiliated Hospital of Medical College, Anhui University of Science & TechnologyHuainan 232001, P.R. China
| | - Suping Qian
- Department of Respiration, Tumour Hospital of Affiliated Huainan Oriental Hospital Group, Anhui University of Science and TechnologyHuainan 232035, P.R. China
| | - Yingru Xin
- Department of Respiration, Tumour Hospital of Affiliated Huainan Oriental Hospital Group, Anhui University of Science and TechnologyHuainan 232035, P.R. China
| | - Yi Gao
- Huainan First People’s Hospital and First Affiliated Hospital of Medical College, Anhui University of Science & TechnologyHuainan 232001, P.R. China
| | - Yongqiang Zhu
- Department of Medical Genetics, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, P.R. China
| | - Xiaolong Tang
- Stem Cell Engineering Research Center, School of Medical, Anhui University of Science & TechnologyHuainan 232001, P.R. China
| |
Collapse
|
27
|
Lorenzo PI, Juárez-Vicente F, Cobo-Vuilleumier N, García-Domínguez M, Gauthier BR. The Diabetes-Linked Transcription Factor PAX4: From Gene to Functional Consequences. Genes (Basel) 2017; 8:genes8030101. [PMID: 28282933 PMCID: PMC5368705 DOI: 10.3390/genes8030101] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 02/24/2017] [Accepted: 03/03/2017] [Indexed: 12/26/2022] Open
Abstract
Paired box 4 (PAX4) is a key factor in the generation of insulin producing β-cells during embryonic development. In adult islets, PAX4 expression is sequestered to a subset of β-cells that are prone to proliferation and more resistant to stress-induced apoptosis. The importance of this transcription factor for adequate pancreatic islets functionality has been manifested by the association of mutations in PAX4 with the development of diabetes, independently of its etiology. Overexpression of this factor in adult islets stimulates β-cell proliferation and increases their resistance to apoptosis. Additionally, in an experimental model of autoimmune diabetes, a novel immunomodulatory function for this factor has been suggested. Altogether these data pinpoint at PAX4 as an important target for novel regenerative therapies for diabetes treatment, aiming at the preservation of the remaining β-cells in parallel to the stimulation of their proliferation to replenish the β-cell mass lost during the progression of the disease. However, the adequate development of such therapies requires the knowledge of the molecular mechanisms controlling the expression of PAX4 as well as the downstream effectors that could account for PAX4 action.
Collapse
Affiliation(s)
- Petra I Lorenzo
- Pancreatic Islet Development and Regeneration Unit, Department of Cell Regeneration and Advanced Therapies, CABIMER (Junta de Andalucía-CSIC-Universidad de Sevilla-Universidad Pablo de Olavide), Calle Américo Vespucio, 24, 41092 Sevilla, Spain.
| | - Francisco Juárez-Vicente
- Cell differentiation Lab, Department of Cell Signaling and Dynamics, CABIMER (Junta de Andalucía-CSIC-Universidad de Sevilla-Universidad Pablo de Olavide), Calle Américo Vespucio, 24, 41092 Sevilla, Spain.
| | - Nadia Cobo-Vuilleumier
- Pancreatic Islet Development and Regeneration Unit, Department of Cell Regeneration and Advanced Therapies, CABIMER (Junta de Andalucía-CSIC-Universidad de Sevilla-Universidad Pablo de Olavide), Calle Américo Vespucio, 24, 41092 Sevilla, Spain.
| | - Mario García-Domínguez
- Cell differentiation Lab, Department of Cell Signaling and Dynamics, CABIMER (Junta de Andalucía-CSIC-Universidad de Sevilla-Universidad Pablo de Olavide), Calle Américo Vespucio, 24, 41092 Sevilla, Spain.
| | - Benoit R Gauthier
- Pancreatic Islet Development and Regeneration Unit, Department of Cell Regeneration and Advanced Therapies, CABIMER (Junta de Andalucía-CSIC-Universidad de Sevilla-Universidad Pablo de Olavide), Calle Américo Vespucio, 24, 41092 Sevilla, Spain.
| |
Collapse
|
28
|
Cheung CYY, Tang CS, Xu A, Lee CH, Au KW, Xu L, Fong CHY, Kwok KHM, Chow WS, Woo YC, Yuen MMA, Hai JSH, Jin YL, Cheung BMY, Tan KCB, Cherny SS, Zhu F, Zhu T, Thomas GN, Cheng KK, Jiang CQ, Lam TH, Tse HF, Sham PC, Lam KSL. Exome-chip association analysis reveals an Asian-specific missense variant in PAX4 associated with type 2 diabetes in Chinese individuals. Diabetologia 2017; 60:107-115. [PMID: 27744525 DOI: 10.1007/s00125-016-4132-z] [Citation(s) in RCA: 18] [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: 05/04/2016] [Accepted: 09/21/2016] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS Genome-wide association studies (GWASs) have identified many common type 2 diabetes-associated variants, mostly at the intronic or intergenic regions. Recent advancements of exome-array genotyping platforms have opened up a novel means for detecting the associations of low-frequency or rare coding variants with type 2 diabetes. We conducted an exomechip association analysis to identify additional type 2 diabetes susceptibility variants in the Chinese population. METHODS An exome-chip association study was conducted by genotyping 5640 Chinese individuals from Hong Kong, using a custom designed exome array, the Asian Exomechip. Single variant association analysis was conducted on 77,468 single nucleotide polymorphisms (SNPs). Fifteen SNPs were subsequently genotyped for replication analysis in an independent Chinese cohort comprising 12,362 individuals from Guangzhou. A combined analysis involving 7189 cases and 10,813 controls was performed. RESULTS In the discovery stage, an Asian-specific coding variant rs2233580 (p.Arg192His) in PAX4, and two variants at the known loci, CDKN2B-AS1 and KCNQ1, were significantly associated with type 2 diabetes with exome-wide significance (p discovery < 6.45 × 10-7). The risk allele (T) of PAX4 rs2233580 was associated with a younger age at diabetes diagnosis. This variant was replicated in an independent cohort and demonstrated a stronger association that reached genome-wide significance (p meta-analysis [p meta] = 3.74 × 10-15) in the combined analysis. CONCLUSIONS/INTERPRETATION We identified the association of a PAX4 Asian-specific missense variant rs2233580 with type 2 diabetes in an exome-chip association analysis, supporting the involvement of PAX4 in the pathogenesis of type 2 diabetes. Our findings suggest PAX4 is a possible effector gene of the 7q32 locus, previously identified from GWAS in Asians.
Collapse
Affiliation(s)
- Chloe Y Y Cheung
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, People's Republic of China
| | - Clara S Tang
- Department of Surgery, University of Hong Kong, Hong Kong, People's Republic of China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, People's Republic of China
- Research Centre of Heart, Brain, Hormone and Healthy Ageing, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, People's Republic of China
- Department of Pharmacology & Pharmacy, University of Hong Kong, Hong Kong, People's Republic of China
| | - Chi-Ho Lee
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, People's Republic of China
| | - Ka-Wing Au
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, People's Republic of China
| | - Lin Xu
- School of Public Health, Room 505, Faculty of Medicine Building, William M.W. Mong Block, University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong, People's Republic of China
| | - Carol H Y Fong
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, People's Republic of China
| | - Kelvin H M Kwok
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, People's Republic of China
| | - Wing-Sun Chow
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, People's Republic of China
| | - Yu-Cho Woo
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, People's Republic of China
| | - Michele M A Yuen
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, People's Republic of China
| | - JoJo S H Hai
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, People's Republic of China
| | - Ya-Li Jin
- Molecular Epidemiological Research Centre, Guangzhou Number 12 Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Bernard M Y Cheung
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, People's Republic of China
| | - Kathryn C B Tan
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, People's Republic of China
| | - Stacey S Cherny
- Department of Psychiatry, University of Hong Kong, Hong Kong, People's Republic of China
| | - Feng Zhu
- Molecular Epidemiological Research Centre, Guangzhou Number 12 Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Tong Zhu
- Molecular Epidemiological Research Centre, Guangzhou Number 12 Hospital, Guangzhou, Guangdong, People's Republic of China
| | - G Neil Thomas
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Kar-Keung Cheng
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Chao-Qiang Jiang
- Molecular Epidemiological Research Centre, Guangzhou Number 12 Hospital, Guangzhou, Guangdong, People's Republic of China
| | - Tai-Hing Lam
- School of Public Health, Room 505, Faculty of Medicine Building, William M.W. Mong Block, University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong, People's Republic of China.
- Molecular Epidemiological Research Centre, Guangzhou Number 12 Hospital, Guangzhou, Guangdong, People's Republic of China.
| | - Hung-Fat Tse
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, People's Republic of China.
- Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, University of Hong Kong, Hong Kong, People's Republic of China.
| | - Pak-Chung Sham
- Department of Psychiatry, University of Hong Kong, Hong Kong, People's Republic of China.
- Centre for Genomic Sciences, Centre for Genomic Sciences, University of Hong Kong, 6/F, HKJC Building for Interdisciplinary Research, 5 Sassoon Road, Pokfulam, Hong Kong, People's Republic of China.
- State Key Laboratory in Brain and Cognitive Sciences, University of Hong Kong, Hong Kong, People's Republic of China.
| | - Karen S L Lam
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Hong Kong, People's Republic of China.
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, People's Republic of China.
- Research Centre of Heart, Brain, Hormone and Healthy Ageing, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, People's Republic of China.
| |
Collapse
|
29
|
Arregi I, Climent M, Iliev D, Strasser J, Gouignard N, Johansson JK, Singh T, Mazur M, Semb H, Artner I, Minichiello L, Pera EM. Retinol Dehydrogenase-10 Regulates Pancreas Organogenesis and Endocrine Cell Differentiation via Paracrine Retinoic Acid Signaling. Endocrinology 2016; 157:4615-4631. [PMID: 27740873 DOI: 10.1210/en.2016-1745] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Vitamin A-derived retinoic acid (RA) signals are critical for the development of several organs, including the pancreas. However, the tissue-specific control of RA synthesis in organ and cell lineage development has only poorly been addressed in vivo. Here, we show that retinol dehydrogenase-10 (Rdh10), a key enzyme in embryonic RA production, has important functions in pancreas organogenesis and endocrine cell differentiation. Rdh10 was expressed in the developing pancreas epithelium and surrounding mesenchyme. Rdh10 null mutant mouse embryos exhibited dorsal pancreas agenesis and a hypoplastic ventral pancreas with retarded tubulogenesis and branching. Conditional disruption of Rdh10 from the endoderm caused increased mortality, reduced body weight, and lowered blood glucose levels after birth. Endodermal Rdh10 deficiency led to a smaller dorsal pancreas with a reduced density of early glucagon+ and insulin+ cells. During the secondary transition, the reduction of Neurogenin3+ endocrine progenitors in the mutant dorsal pancreas accounted for fewer α- and β-cells. Changes in the expression of α- and β-cell-specific transcription factors indicated that Rdh10 might also participate in the terminal differentiation of endocrine cells. Together, our results highlight the importance of both mesenchymal and epithelial Rdh10 for pancreogenesis and the first wave of endocrine cell differentiation. We further propose a model in which the Rdh10-expressing exocrine tissue acts as an essential source of RA signals in the second wave of endocrine cell differentiation.
Collapse
Affiliation(s)
- Igor Arregi
- Lund Stem Cell Center (I.Arr., M.C., D.I., J.S., N.G., J.K.J., T.S., M.M., I.Art., E.M.P.), Lund University, SE-22184 Lund, Sweden; The Danish Stem Cell Center (H.S.), University of Copenhagen, DK-2200 Copenhagen, Denmark; and Department of Pharmacology (L.M.), University of Oxford, OX1 3QT Oxford, United Kingdom
| | - Maria Climent
- Lund Stem Cell Center (I.Arr., M.C., D.I., J.S., N.G., J.K.J., T.S., M.M., I.Art., E.M.P.), Lund University, SE-22184 Lund, Sweden; The Danish Stem Cell Center (H.S.), University of Copenhagen, DK-2200 Copenhagen, Denmark; and Department of Pharmacology (L.M.), University of Oxford, OX1 3QT Oxford, United Kingdom
| | - Dobromir Iliev
- Lund Stem Cell Center (I.Arr., M.C., D.I., J.S., N.G., J.K.J., T.S., M.M., I.Art., E.M.P.), Lund University, SE-22184 Lund, Sweden; The Danish Stem Cell Center (H.S.), University of Copenhagen, DK-2200 Copenhagen, Denmark; and Department of Pharmacology (L.M.), University of Oxford, OX1 3QT Oxford, United Kingdom
| | - Jürgen Strasser
- Lund Stem Cell Center (I.Arr., M.C., D.I., J.S., N.G., J.K.J., T.S., M.M., I.Art., E.M.P.), Lund University, SE-22184 Lund, Sweden; The Danish Stem Cell Center (H.S.), University of Copenhagen, DK-2200 Copenhagen, Denmark; and Department of Pharmacology (L.M.), University of Oxford, OX1 3QT Oxford, United Kingdom
| | - Nadège Gouignard
- Lund Stem Cell Center (I.Arr., M.C., D.I., J.S., N.G., J.K.J., T.S., M.M., I.Art., E.M.P.), Lund University, SE-22184 Lund, Sweden; The Danish Stem Cell Center (H.S.), University of Copenhagen, DK-2200 Copenhagen, Denmark; and Department of Pharmacology (L.M.), University of Oxford, OX1 3QT Oxford, United Kingdom
| | - Jenny K Johansson
- Lund Stem Cell Center (I.Arr., M.C., D.I., J.S., N.G., J.K.J., T.S., M.M., I.Art., E.M.P.), Lund University, SE-22184 Lund, Sweden; The Danish Stem Cell Center (H.S.), University of Copenhagen, DK-2200 Copenhagen, Denmark; and Department of Pharmacology (L.M.), University of Oxford, OX1 3QT Oxford, United Kingdom
| | - Tania Singh
- Lund Stem Cell Center (I.Arr., M.C., D.I., J.S., N.G., J.K.J., T.S., M.M., I.Art., E.M.P.), Lund University, SE-22184 Lund, Sweden; The Danish Stem Cell Center (H.S.), University of Copenhagen, DK-2200 Copenhagen, Denmark; and Department of Pharmacology (L.M.), University of Oxford, OX1 3QT Oxford, United Kingdom
| | - Magdalena Mazur
- Lund Stem Cell Center (I.Arr., M.C., D.I., J.S., N.G., J.K.J., T.S., M.M., I.Art., E.M.P.), Lund University, SE-22184 Lund, Sweden; The Danish Stem Cell Center (H.S.), University of Copenhagen, DK-2200 Copenhagen, Denmark; and Department of Pharmacology (L.M.), University of Oxford, OX1 3QT Oxford, United Kingdom
| | - Henrik Semb
- Lund Stem Cell Center (I.Arr., M.C., D.I., J.S., N.G., J.K.J., T.S., M.M., I.Art., E.M.P.), Lund University, SE-22184 Lund, Sweden; The Danish Stem Cell Center (H.S.), University of Copenhagen, DK-2200 Copenhagen, Denmark; and Department of Pharmacology (L.M.), University of Oxford, OX1 3QT Oxford, United Kingdom
| | - Isabella Artner
- Lund Stem Cell Center (I.Arr., M.C., D.I., J.S., N.G., J.K.J., T.S., M.M., I.Art., E.M.P.), Lund University, SE-22184 Lund, Sweden; The Danish Stem Cell Center (H.S.), University of Copenhagen, DK-2200 Copenhagen, Denmark; and Department of Pharmacology (L.M.), University of Oxford, OX1 3QT Oxford, United Kingdom
| | - Liliana Minichiello
- Lund Stem Cell Center (I.Arr., M.C., D.I., J.S., N.G., J.K.J., T.S., M.M., I.Art., E.M.P.), Lund University, SE-22184 Lund, Sweden; The Danish Stem Cell Center (H.S.), University of Copenhagen, DK-2200 Copenhagen, Denmark; and Department of Pharmacology (L.M.), University of Oxford, OX1 3QT Oxford, United Kingdom
| | - Edgar M Pera
- Lund Stem Cell Center (I.Arr., M.C., D.I., J.S., N.G., J.K.J., T.S., M.M., I.Art., E.M.P.), Lund University, SE-22184 Lund, Sweden; The Danish Stem Cell Center (H.S.), University of Copenhagen, DK-2200 Copenhagen, Denmark; and Department of Pharmacology (L.M.), University of Oxford, OX1 3QT Oxford, United Kingdom
| |
Collapse
|
30
|
PAX4 R192H and P321H polymorphisms in type 2 diabetes and their functional defects. J Hum Genet 2016; 61:943-949. [PMID: 27334367 DOI: 10.1038/jhg.2016.80] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 05/30/2016] [Accepted: 05/31/2016] [Indexed: 01/05/2023]
Abstract
We have previously identified PAX4 mutations causing MODY9 and a recent genome-wide association study reported a susceptibility locus of type 2 diabetes (T2D) near PAX4. In this study, we aim to investigate the association between PAX4 polymorphisms and T2D in Thai patients and examine functions of PAX4 variant proteins. PAX4 rs2233580 (R192H) and rs712701 (P321H) were genotyped in 746 patients with T2D and 562 healthy normal control subjects by PCR and restriction-fragment length polymorphism method. PAX4 variant proteins were investigated for repressor function on human insulin and glucagon promoters and for cell viability and apoptosis upon high glucose exposure. Genotype and allele frequencies of PAX4 rs2233580 were more frequent in patients with T2D than in control subjects (P=0.001 and 0.0006, respectively) with odds ratio of 1.66 (P=0.001; 95% confidence interval, 1.22-2.27). PAX4 rs712701 was not associated with T2D but it was in linkage disequilibrium with rs2233580. The 192H/321H (A/A) haplotype was more frequent in T2D patients than in controls (9.5% vs 6.6%; P=0.009). PAX4 R192H, but not PAX4 P321H, impaired repression activities on insulin and glucagon promoters and decreased transcript levels of genes required to maintain β-cell function, proliferation and survival. Viability of β-cell was reduced under glucotoxic stress condition for the cells overexpressing either PAX4 R192H or PAX4 P321H or both. Thus these PAX4 polymorphisms may increase T2D risk by defective transcription regulation of target genes and/or decreased β-cell survival in high glucose condition.
Collapse
|
31
|
Shaer A, Azarpira N, Karimi MH, Soleimani M, Dehghan S. Differentiation of Human-Induced Pluripotent Stem Cells Into Insulin-Producing Clusters by MicroRNA-7. EXP CLIN TRANSPLANT 2016; 14:555-563. [PMID: 26103160 DOI: 10.6002/ect.2014.0144] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Diabetes results from inadequate insulin production from pancreatic β-cells. Islet cell replacement is an effective approach for diabetes treatment; however, it is not sufficient for all diabetic patients. Thus, finding a new source with effective maturation of β-cells is the major goal of many studies. MicroRNAs are a class of small noncoding ribonucleic acid that regulate gene expression through posttranscriptional mechanisms. MicroRNA-7 has high expression level during pancreatic islet development in humans, thereby playing a critical role in pancreatic β-cell function. We study aimed to develop a protocol to differentiate human-induced pluripotent stem cells efficiently into isletlike cell clusters in vitro by using microRNA-7. MATERIALS AND METHODS Human-induced pluripotent stem cell colonies were transfected with hsa-microRNA-7 by using siPORT NeoFX transfection agent. Total ribonucleic acid was extracted 24 and 48 hours after transfection. The expression of transcription factors which were important during pancreases development was also performed. On the third day, the potency of the clusters was assessed in response to high glucose levels. Diphenylthiocarbazone was used to identify the existence of the β-cells. The presence of insulin and Neurogenin-3 proteins was investigated by immunocytochemistry. RESULTS Morphologic changes were observed on the first day after chemical transfection, and cell clusters were formed on the third day. The expression of pancreatic specific transcription factors was increased on the first day and significantly increased on the second day. The isletlike cell clusters were positive for insulin and Neurogenin-3 proteins in immunocytochemistry. The clusters were stained with Diphenylthiocarbazone and secreted insulin in a glucose challenge test. CONCLUSIONS MicroRNA-7 transcription factor network is important in pancreatic endocrine differentiation. Chemical transfection with microRNA-7 can differentiate human induced pluripotent stem cells into functional isletlike cell clusters in a short time.
Collapse
Affiliation(s)
- Anahita Shaer
- From the Department of Genetics, Zarghan Branch, Islamic Azad University, Fars, Iran; and Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | | | |
Collapse
|
32
|
Yang Y, Chan L. Monogenic Diabetes: What It Teaches Us on the Common Forms of Type 1 and Type 2 Diabetes. Endocr Rev 2016; 37:190-222. [PMID: 27035557 PMCID: PMC4890265 DOI: 10.1210/er.2015-1116] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To date, more than 30 genes have been linked to monogenic diabetes. Candidate gene and genome-wide association studies have identified > 50 susceptibility loci for common type 1 diabetes (T1D) and approximately 100 susceptibility loci for type 2 diabetes (T2D). About 1-5% of all cases of diabetes result from single-gene mutations and are called monogenic diabetes. Here, we review the pathophysiological basis of the role of monogenic diabetes genes that have also been found to be associated with common T1D and/or T2D. Variants of approximately one-third of monogenic diabetes genes are associated with T2D, but not T1D. Two of the T2D-associated monogenic diabetes genes-potassium inward-rectifying channel, subfamily J, member 11 (KCNJ11), which controls glucose-stimulated insulin secretion in the β-cell; and peroxisome proliferator-activated receptor γ (PPARG), which impacts multiple tissue targets in relation to inflammation and insulin sensitivity-have been developed as major antidiabetic drug targets. Another monogenic diabetes gene, the preproinsulin gene (INS), is unique in that INS mutations can cause hyperinsulinemia, hyperproinsulinemia, neonatal diabetes mellitus, one type of maturity-onset diabetes of the young (MODY10), and autoantibody-negative T1D. Dominant heterozygous INS mutations are the second most common cause of permanent neonatal diabetes. Moreover, INS gene variants are strongly associated with common T1D (type 1a), but inconsistently with T2D. Variants of the monogenic diabetes gene Gli-similar 3 (GLIS3) are associated with both T1D and T2D. GLIS3 is a key transcription factor in insulin production and β-cell differentiation during embryonic development, which perturbation forms the basis of monogenic diabetes as well as its association with T1D. GLIS3 is also required for compensatory β-cell proliferation in adults; impairment of this function predisposes to T2D. Thus, monogenic forms of diabetes are invaluable "human models" that have contributed to our understanding of the pathophysiological basis of common T1D and T2D.
Collapse
Affiliation(s)
- Yisheng Yang
- Division of Endocrinology (Y.Y.), Department of Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio 44109; and Diabetes and Endocrinology Research Center (L.C.), Division of Diabetes, Endocrinology and Metabolism, Departments of Medicine, Molecular and Cellular Biology, Biochemistry and Molecular Biology, and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Lawrence Chan
- Division of Endocrinology (Y.Y.), Department of Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio 44109; and Diabetes and Endocrinology Research Center (L.C.), Division of Diabetes, Endocrinology and Metabolism, Departments of Medicine, Molecular and Cellular Biology, Biochemistry and Molecular Biology, and Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
33
|
Abstract
Type two diabetes (T2D) is a challenging metabolic disorder for which a cure has not yet been found. Its etiology is associated with several phenomena, including significant loss of insulin-producing, beta cell (β cell) mass via progressive programmed cell death and disrupted cellular autophagy. In diabetes, the etiology of β cell death and the role of mitochondria are complex and involve several layers of mechanisms. Understanding the dynamics of those mechanisms could permit researchers to develop an intervention for the progressive loss of β cells. Currently, diabetes research has shifted toward rejuvenation and plasticity technology and away from the simplified approach of hormonal compensation. Diabetes research is currently challenged by questions such as how to enhance cell survival, decrease apoptosis and replenish β cell mass in diabetic patients. In this review, we discuss evidence that β cell development and mass formation are guided by specific signaling systems, particularly hormones, transcription factors, and growth factors, all of which could be manipulated to enhance mass growth. There is also strong evidence that β cells are dynamically active cells, which, under specific conditions such as obesity, can increase in size and subsequently increase insulin secretion. In certain cases of aggressive or advanced forms of T2D, β cells become markedly impaired, and the only alternatives for maintaining glucose homeostasis are through partial or complete cell grafting (the Edmonton protocol). In these cases, the harvesting of an enriched population of viable β cells is required for transplantation. This task necessitates a deep understanding of the pharmacological agents that affect β cell survival, mass, and function. The aim of this review is to initiate discussion about the important signals in pancreatic β cell development and mass formation and to highlight the process by which cell death occurs in diabetes. This review also examines the attempts that have been made to recover or increase cell mass in diabetic patients by using various pharmacological agents.
Collapse
Affiliation(s)
- Husnia I Marrif
- Department of Pharmacology, Faculty of Medicine, University of Benghazi Benghazi, Libya
| | - Salma I Al-Sunousi
- Department of Histology and Anatomy, Faculty of Medicine, University of Benghazi Benghazi, Libya
| |
Collapse
|
34
|
Sujjitjoon J, Kooptiwut S, Chongjaroen N, Tangjittipokin W, Plengvidhya N, Yenchitsomanus PT. Aberrant mRNA splicing of paired box 4 (PAX4) IVS7-1G>A mutation causing maturity-onset diabetes of the young, type 9. Acta Diabetol 2016; 53:205-16. [PMID: 25951767 DOI: 10.1007/s00592-015-0760-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 04/12/2015] [Indexed: 01/24/2023]
Abstract
AIMS Paired box 4 (PAX4) mutations cause maturity-onset diabetes of the young, type 9 (MODY9). The molecular defect and alteration of PAX4 function associated with the mutation PAX4 IVS7-1G>A in a family with MODY9 and severe diabetic complications were studied. METHODS We investigated the functional consequences of PAX4 IVS7-1G>A on mRNA splicing using minigene assays. Wild-type and mutant PAX4 were expressed in mouse pancreatic β- and α-cell lines, and protein levels and translocation of PAX4 into the nucleus were determined. We also examined transcriptional repression of PAX4 target-gene promoters and β-cell viability under diabetic-like (high-glucose) conditions. RESULTS PAX4 IVS7-1G>A disrupts an acceptor splice site, causing an adjacent cryptic splice site within exon 8 to be used, resulting in a three-nucleotide deletion and glutamine deletion at position 250 (p.Q250del). Wild-type and PAX4 Q250del proteins were expressed at similar levels and could translocate normally into the nucleus in βTC3 and αTC1.9 cells. However, the repressor functions of PAX4 Q250del on human insulin and glucagon promoters in INS-1 832/13 and αTC1.9 cells were significantly decreased, compared with that of wild-type PAX4. Moreover, the rate of apoptosis was increased in INS-1 cells over-expressing PAX4 Q250del when cultured in high-glucose conditions. CONCLUSIONS PAX4 IVS7-1G>A caused aberrant mRNA splicing and PAX4 Q250 deletion. The mutation impaired PAX4 repressor functions on target-gene promoters and increased susceptibility to apoptosis upon high glucose exposure. Thus, PAX4 IVS7-1G>A contributes to the pathogenesis of diabetes in this MODY9 family through β-cell dysfunction.
Collapse
Affiliation(s)
- Jatuporn Sujjitjoon
- Division of Molecular Medicine, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Suwattanee Kooptiwut
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Nalinee Chongjaroen
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Watip Tangjittipokin
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Nattachet Plengvidhya
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| | - Pa-Thai Yenchitsomanus
- Division of Molecular Medicine, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
35
|
Adaptive methylation regulation of p53 pathway in sympatric speciation of blind mole rats, Spalax. Proc Natl Acad Sci U S A 2016; 113:2146-51. [PMID: 26858405 DOI: 10.1073/pnas.1522658112] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Epigenetic modifications play significant roles in adaptive evolution. The tumor suppressor p53, well known for controlling cell fate and maintaining genomic stability, is much less known as a master gene in environmental adaptation involving methylation modifications. The blind subterranean mole rat Spalax eherenbergi superspecies in Israel consists of four species that speciated peripatrically. Remarkably, the northern Galilee species Spalax galili (2n = 52) underwent adaptive ecological sympatric speciation, caused by the sharply divergent chalk and basalt ecologies. This was demonstrated by mitochondrial and nuclear genomic evidence. Here we show that the expression patterns of the p53 regulatory pathway diversified between the abutting sympatric populations of S. galili in sharply divergent chalk-basalt ecologies. We identified higher methylation on several sites of the p53 promoter in the population living in chalk soil (chalk population). Site mutagenesis showed that methylation on these sites linked to the transcriptional repression of p53 involving Cut-Like Homeobox 1 (Cux1), paired box 4 (Pax 4), Pax 6, and activator protein 1 (AP-1). Diverse expression levels of p53 between the incipiently sympatrically speciating chalk-basalt abutting populations of S. galili selectively affected cell-cycle arrest but not apoptosis. We hypothesize that methylation modification of p53 has adaptively shifted in supervising its target genes during sympatric speciation of S. galili to cope with the contrasting environmental stresses of the abutting divergent chalk-basalt ecologies.
Collapse
|
36
|
Voevoda MI, Ivanova AA, Shakhtshneider EV, Ovsyannikova AK, Mikhailova SV, Astrakova KS, Voevoda SM, Rymar OD. Molecular genetics of maturity-onset diabetes of the young. TERAPEVT ARKH 2016. [DOI: 10.17116/terarkh2016884117-124] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
37
|
Abstract
Lineage tracing studies have revealed that transcription factors play a cardinal role in pancreatic development, differentiation and function. Three transitions define pancreatic organogenesis, differentiation and maturation. In the primary transition, when pancreatic organogenesis is initiated, there is active proliferation of pancreatic progenitor cells. During the secondary transition, defined by differentiation, there is growth, branching, differentiation and pancreatic cell lineage allocation. The tertiary transition is characterized by differentiated pancreatic cells that undergo further remodeling, including apoptosis, replication and neogenesis thereby establishing a mature organ. Transcription factors function at multiple levels and may regulate one another and auto-regulate. The interaction between extrinsic signals from non-pancreatic tissues and intrinsic transcription factors form a complex gene regulatory network ultimately culminating in the different cell lineages and tissue types in the developing pancreas. Mutations in these transcription factors clinically manifest as subtypes of diabetes mellitus. Current treatment for diabetes is not curative and thus, developmental biologists and stem cell researchers are utilizing knowledge of normal pancreatic development to explore novel therapeutic alternatives. This review summarizes current knowledge of transcription factors involved in pancreatic development and β-cell differentiation in rodents.
Collapse
Affiliation(s)
- Reshmi Dassaye
- a Discipline of Pharmaceutical Sciences; Nelson R. Mandela School of Medicine, University of KwaZulu-Natal , Durban , South Africa
| | - Strini Naidoo
- a Discipline of Pharmaceutical Sciences; Nelson R. Mandela School of Medicine, University of KwaZulu-Natal , Durban , South Africa
| | - Marlon E Cerf
- b Diabetes Discovery Platform, South African Medical Research Council , Cape Town , South Africa
| |
Collapse
|
38
|
PAX4 Gene Transfer Induces α-to-β Cell Phenotypic Conversion and Confers Therapeutic Benefits for Diabetes Treatment. Mol Ther 2015; 24:251-260. [PMID: 26435408 DOI: 10.1038/mt.2015.181] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 09/25/2015] [Indexed: 12/19/2022] Open
Abstract
The transcription factor Pax4 plays a critical role in the determination of α- versus β-cell lineage during endocrine pancreas development. In this study, we explored whether Pax4 gene transfer into α-cells could convert them into functional β-cells and thus provide therapeutic benefits for insulin-deficient diabetes. We found that Pax4 delivered by adenoviral vector, Ad5.Pax4, induced insulin expression and reduced glucagon expression in αTC1.9 cells. More importantly, these cells exhibited glucose-stimulated insulin secretion, a key feature of functional β-cells. When injected into streptozotocin-induced diabetic mice, Pax4-treated αTC1.9 cells significantly reduced blood glucose, and the mice showed better glucose tolerance, supporting that Pax4 gene transfer into αTC1.9 cells resulted in the formation of functional β-cells. Furthermore, treatment of primary human islets with Ad5.Pax4 resulted in significantly improved β-cell function. Detection of glucagon(+)/Pax4(+)/Insulin(+) cells argued for Pax4-induced α-to-β cell transitioning. This was further supported by quantification of glucagon and insulin bi-hormonal cells, which was significantly higher in Pax4-treated islets than in controls. Finally, direct administration of Ad5.Pax4 into the pancreas of insulin-deficient mice ameliorated hyperglycemia. Taken together, our data demonstrate that manipulating Pax4 gene expression represents a viable therapeutic strategy for the treatment of insulin deficient diabetes.
Collapse
|
39
|
Origins and evolvability of the PAX family. Semin Cell Dev Biol 2015; 44:64-74. [DOI: 10.1016/j.semcdb.2015.08.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 08/07/2015] [Accepted: 08/22/2015] [Indexed: 01/18/2023]
|
40
|
Mayran A, Pelletier A, Drouin J. Pax factors in transcription and epigenetic remodelling. Semin Cell Dev Biol 2015; 44:135-44. [PMID: 26234816 DOI: 10.1016/j.semcdb.2015.07.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/22/2015] [Accepted: 07/24/2015] [Indexed: 11/25/2022]
Abstract
The nine Pax transcription factors that constitute the mammalian family of paired domain (PD) factors play key roles in many developmental processes. As DNA binding transcription factors, they exhibit tremendous variability and complexity in their DNA recognition patterns. This is ascribed to the presence of multiple DNA binding structural domains, namely helix-turn-helix (HTH) domains. The PD contains two HTH subdomains and four of the nine Pax factors have an additional HTH domain, the homeodomain (HD). We now review these diverse DNA binding modalities together with their properties as transcriptional activators and repressors. The action of Pax factors on gene expression is also exerted through recruitment of chromatin remodelling complexes that introduce either activating or repressive chromatin marks. Interestingly, the recent demonstration that Pax7 has pioneer activity, the unique property to "open" chromatin, further underlines the mechanistic versatility and the developmental importance of these factors.
Collapse
Affiliation(s)
- Alexandre Mayran
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
| | - Audrey Pelletier
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada
| | - Jacques Drouin
- Laboratoire de Génétique Moléculaire, Institut de Recherches Cliniques de Montréal (IRCM), Montréal, QC H2W 1R7, Canada.
| |
Collapse
|
41
|
van der Meulen T, Huising MO. Role of transcription factors in the transdifferentiation of pancreatic islet cells. J Mol Endocrinol 2015; 54:R103-17. [PMID: 25791577 PMCID: PMC4373662 DOI: 10.1530/jme-14-0290] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The α and β cells act in concert to maintain blood glucose. The α cells release glucagon in response to low levels of glucose to stimulate glycogenolysis in the liver. In contrast, β cells release insulin in response to elevated levels of glucose to stimulate peripheral glucose disposal. Despite these opposing roles in glucose homeostasis, α and β cells are derived from a common progenitor and share many proteins important for glucose sensing and hormone secretion. Results from recent work have underlined these similarities between the two cell types by revealing that β-to-α as well as α-to-β transdifferentiation can take place under certain experimental circumstances. These exciting findings highlight unexpected plasticity of adult islets and offer hope of novel therapeutic paths to replenish β cells in diabetes. In this review, we focus on the transcription factor networks that establish and maintain pancreatic endocrine cell identity and how they may be perturbed to facilitate transdifferentiation.
Collapse
Affiliation(s)
- Talitha van der Meulen
- Department of NeurobiologyPhysiology and Behavior, College of Biological SciencesDepartment of Physiology and Membrane BiologySchool of Medicine, University of California, 193 Briggs Hall, One Shields Avenue, Davis, California 95616, USA
| | - Mark O Huising
- Department of NeurobiologyPhysiology and Behavior, College of Biological SciencesDepartment of Physiology and Membrane BiologySchool of Medicine, University of California, 193 Briggs Hall, One Shields Avenue, Davis, California 95616, USA Department of NeurobiologyPhysiology and Behavior, College of Biological SciencesDepartment of Physiology and Membrane BiologySchool of Medicine, University of California, 193 Briggs Hall, One Shields Avenue, Davis, California 95616, USA
| |
Collapse
|
42
|
Kaneto H, Matsuoka TA. Role of pancreatic transcription factors in maintenance of mature β-cell function. Int J Mol Sci 2015; 16:6281-97. [PMID: 25794287 PMCID: PMC4394532 DOI: 10.3390/ijms16036281] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/10/2015] [Accepted: 02/16/2015] [Indexed: 12/12/2022] Open
Abstract
A variety of pancreatic transcription factors including PDX-1 and MafA play crucial roles in the pancreas and function for the maintenance of mature β-cell function. However, when β-cells are chronically exposed to hyperglycemia, expression and/or activities of such transcription factors are reduced, which leads to deterioration of β-cell function. These phenomena are well known as β-cell glucose toxicity in practical medicine as well as in the islet biology research area. Here we describe the possible mechanism for β-cell glucose toxicity found in type 2 diabetes. It is likely that reduced expression levels of PDX-1 and MafA lead to suppression of insulin biosynthesis and secretion. In addition, expression levels of incretin receptors (GLP-1 and GIP receptors) in β-cells are decreased, which likely contributes to the impaired incretin effects found in diabetes. Taken together, down-regulation of insulin gene transcription factors and incretin receptors explains, at least in part, the molecular mechanism for β-cell glucose toxicity.
Collapse
Affiliation(s)
- Hideaki Kaneto
- Department of Diabetes, Endocrinology and Metabolism, Kawasaki Medical School, 577, Matsushima, Kurashiki 701-0192, Japan.
| | - Taka-aki Matsuoka
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan.
| |
Collapse
|
43
|
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: 221] [Impact Index Per Article: 20.1] [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
|
44
|
PAX genes in childhood oncogenesis: developmental biology gone awry? Oncogene 2014; 34:2681-9. [PMID: 25043308 DOI: 10.1038/onc.2014.209] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/10/2014] [Accepted: 06/11/2014] [Indexed: 01/27/2023]
Abstract
Childhood solid tumors often arise from embryonal-like cells, which are distinct from the epithelial cancers observed in adults, and etiologically can be considered as 'developmental patterning gone awry'. Paired-box (PAX) genes encode a family of evolutionarily conserved transcription factors that are important regulators of cell lineage specification, migration and tissue patterning. PAX loss-of-function mutations are well known to cause potent developmental phenotypes in animal models and underlie genetic disease in humans, whereas dysregulation and/or genetic modification of PAX genes have been shown to function as critical triggers for human tumorigenesis. Consequently, exploring PAX-related pathobiology generates insights into both normal developmental biology and key molecular mechanisms that underlie pediatric cancer, which are the topics of this review.
Collapse
|
45
|
Damasceno DC, Netto AO, Iessi IL, Gallego FQ, Corvino SB, Dallaqua B, Sinzato YK, Bueno A, Calderon IMP, Rudge MVC. Streptozotocin-induced diabetes models: pathophysiological mechanisms and fetal outcomes. BIOMED RESEARCH INTERNATIONAL 2014; 2014:819065. [PMID: 24977161 PMCID: PMC4058231 DOI: 10.1155/2014/819065] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/30/2014] [Accepted: 05/14/2014] [Indexed: 12/26/2022]
Abstract
Glucose homeostasis is controlled by endocrine pancreatic cells, and any pancreatic disturbance can result in diabetes. Because 8% to 12% of diabetic pregnant women present with malformed fetuses, there is great interest in understanding the etiology, pathophysiological mechanisms, and treatment of gestational diabetes. Hyperglycemia enhances the production of reactive oxygen species, leading to oxidative stress, which is involved in diabetic teratogenesis. It has also been suggested that maternal diabetes alters embryonic gene expression, which might cause malformations. Due to ethical issues involving human studies that sometimes have invasive aspects and the multiplicity of uncontrolled variables that can alter the uterine environment during clinical studies, it is necessary to use animal models to better understand diabetic pathophysiology. This review aimed to gather information about pathophysiological mechanisms and fetal outcomes in streptozotocin-induced diabetic rats. To understand the pathophysiological mechanisms and factors involved in diabetes, the use of pancreatic regeneration studies is increasing in an attempt to understand the behavior of pancreatic beta cells. In addition, these studies suggest a new preventive concept as a treatment basis for diabetes, introducing therapeutic efforts to minimize or prevent diabetes-induced oxidative stress, DNA damage, and teratogenesis.
Collapse
Affiliation(s)
- D. C. Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Graduate Program in Gynecology, Obstetrics and Mastology, Botucatu Medical School, UNESP-Universidade Estadual Paulista, Distrito de Rubião Júnior S/N, 18618-970 Botucatu, SP, Brazil
- Department of Gynecology and Obstetrics, Botucatu Medical School, UNESP-Univsidade Estadual Paulista, Distrito de Rubião Júnior S/N, 18618-970 Botucatu, SP, Brazil
| | - A. O. Netto
- Laboratory of Experimental Research on Gynecology and Obstetrics, Graduate Program in Gynecology, Obstetrics and Mastology, Botucatu Medical School, UNESP-Universidade Estadual Paulista, Distrito de Rubião Júnior S/N, 18618-970 Botucatu, SP, Brazil
| | - I. L. Iessi
- Laboratory of Experimental Research on Gynecology and Obstetrics, Graduate Program in Gynecology, Obstetrics and Mastology, Botucatu Medical School, UNESP-Universidade Estadual Paulista, Distrito de Rubião Júnior S/N, 18618-970 Botucatu, SP, Brazil
| | - F. Q. Gallego
- Laboratory of Experimental Research on Gynecology and Obstetrics, Graduate Program in Gynecology, Obstetrics and Mastology, Botucatu Medical School, UNESP-Universidade Estadual Paulista, Distrito de Rubião Júnior S/N, 18618-970 Botucatu, SP, Brazil
| | - S. B. Corvino
- Laboratory of Experimental Research on Gynecology and Obstetrics, Graduate Program in Gynecology, Obstetrics and Mastology, Botucatu Medical School, UNESP-Universidade Estadual Paulista, Distrito de Rubião Júnior S/N, 18618-970 Botucatu, SP, Brazil
| | - B. Dallaqua
- Laboratory of Experimental Research on Gynecology and Obstetrics, Graduate Program in Gynecology, Obstetrics and Mastology, Botucatu Medical School, UNESP-Universidade Estadual Paulista, Distrito de Rubião Júnior S/N, 18618-970 Botucatu, SP, Brazil
| | - Y. K. Sinzato
- Laboratory of Experimental Research on Gynecology and Obstetrics, Graduate Program in Gynecology, Obstetrics and Mastology, Botucatu Medical School, UNESP-Universidade Estadual Paulista, Distrito de Rubião Júnior S/N, 18618-970 Botucatu, SP, Brazil
| | - A. Bueno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Graduate Program in Gynecology, Obstetrics and Mastology, Botucatu Medical School, UNESP-Universidade Estadual Paulista, Distrito de Rubião Júnior S/N, 18618-970 Botucatu, SP, Brazil
| | - I. M. P. Calderon
- Laboratory of Experimental Research on Gynecology and Obstetrics, Graduate Program in Gynecology, Obstetrics and Mastology, Botucatu Medical School, UNESP-Universidade Estadual Paulista, Distrito de Rubião Júnior S/N, 18618-970 Botucatu, SP, Brazil
| | - M. V. C. Rudge
- Laboratory of Experimental Research on Gynecology and Obstetrics, Graduate Program in Gynecology, Obstetrics and Mastology, Botucatu Medical School, UNESP-Universidade Estadual Paulista, Distrito de Rubião Júnior S/N, 18618-970 Botucatu, SP, Brazil
| |
Collapse
|
46
|
A common functional regulatory variant at a type 2 diabetes locus upregulates ARAP1 expression in the pancreatic beta cell. Am J Hum Genet 2014; 94:186-97. [PMID: 24439111 DOI: 10.1016/j.ajhg.2013.12.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 12/11/2013] [Indexed: 12/30/2022] Open
Abstract
Genome-wide association studies (GWASs) have identified more than 70 loci associated with type 2 diabetes (T2D), but for most, the underlying causal variants, associated genes, and functional mechanisms remain unknown. At a T2D- and fasting-proinsulin-associated locus on 11q13.4, we have identified a functional regulatory DNA variant, a candidate target gene, and a plausible underlying molecular mechanism. Fine mapping, conditional analyses, and exome array genotyping in 8,635 individuals from the Metabolic Syndrome in Men study confirmed a single major association signal between fasting proinsulin and noncoding variants (p = 7.4 × 10(-50)). Measurement of allele-specific mRNA levels in human pancreatic islet samples heterozygous for rs11603334 showed that the T2D-risk and proinsulin-decreasing allele (C) is associated with increased ARAP1 expression (p < 0.02). We evaluated four candidate functional SNPs for allelic effects on transcriptional activity by performing reporter assays in rodent pancreatic beta cell lines. The C allele of rs11603334, located near one of the ARAP1 promoters, exhibited 2-fold higher transcriptional activity than did the T allele (p < 0.0001); three other candidate SNPs showed no allelic differences. Electrophoretic mobility shift assays demonstrated decreased binding of pancreatic beta cell transcriptional regulators PAX6 and PAX4 to the rs11603334 C allele. Collectively, these data suggest that the T2D-risk allele of rs11603334 could abrogate binding of a complex containing PAX6 and PAX4 and thus lead to increased promoter activity and ARAP1 expression in human pancreatic islets. This work suggests that increased ARAP1 expression might contribute to T2D susceptibility at this GWAS locus.
Collapse
|
47
|
Abstract
Human embryonic stem cells (hESCs) are pluripotent and capable of generating new β-cells, but current in vitro differentiation protocols generally fail to produce mature, glucose-responsive, unihormonal β-cells. Instead, these methods tend to produce immature polyhormonal endocrine cells which mature in vivo into glucagon-positive α-cells. PAX4 is an established transcription factor in β-cell development and function, and is capable of converting glucagon-positive cells to insulin-positive cells in mice. Work in human and mouse ESCs has shown that constitutive PAX4 expression promotes the development of insulin-positive cells, but whether acute PAX4 expression is sufficient to guide specific endocrine cell fates has not been addressed in hESCs. In this study, we applied recombinant adenovirus to ectopically express human PAX4 in hESC-derived pancreatic progenitors, with the aim of influencing the endocrine developmental cascade away from polyhormonal cells toward unihormonal insulin-positive cells. Gene delivery to pancreatic progenitors was efficient and dose-dependent. By the end of in vitro differentiation, PAX4 reduced ARX expression, but only the high dose tested significantly reduced glucagon release. Single cell analysis revealed that while PAX4 did not alter the proportion of endocrine cells, it did reduce the number of glucagon-positive cells and increased the number of unihormonal insulin-positive cells. These data suggest that acute PAX4 overexpression can reduce expression of ARX and glucagon resulting in improved numbers of unihormonal insulin-positive cells.
Collapse
Affiliation(s)
- Blair K Gage
- Department of Cellular and Physiological Sciences; University of British Columbia; Vancouver, BC Canada
| | - Robert K Baker
- Department of Cellular and Physiological Sciences; University of British Columbia; Vancouver, BC Canada
| | - Timothy J Kieffer
- Department of Cellular and Physiological Sciences; University of British Columbia; Vancouver, BC Canada
- Department of Surgery; University of British Columbia; Vancouver, BC Canada
- Correspondence to: Timothy J Kieffer,
| |
Collapse
|
48
|
Brand LH, Henneges C, Schüssler A, Kolukisaoglu HÜ, Koch G, Wallmeroth N, Hecker A, Thurow K, Zell A, Harter K, Wanke D. Screening for protein-DNA interactions by automatable DNA-protein interaction ELISA. PLoS One 2013; 8:e75177. [PMID: 24146751 PMCID: PMC3795721 DOI: 10.1371/journal.pone.0075177] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 08/12/2013] [Indexed: 12/22/2022] Open
Abstract
DNA-binding proteins (DBPs), such as transcription factors, constitute about 10% of the protein-coding genes in eukaryotic genomes and play pivotal roles in the regulation of chromatin structure and gene expression by binding to short stretches of DNA. Despite their number and importance, only for a minor portion of DBPs the binding sequence had been disclosed. Methods that allow the de novo identification of DNA-binding motifs of known DBPs, such as protein binding microarray technology or SELEX, are not yet suited for high-throughput and automation. To close this gap, we report an automatable DNA-protein-interaction (DPI)-ELISA screen of an optimized double-stranded DNA (dsDNA) probe library that allows the high-throughput identification of hexanucleotide DNA-binding motifs. In contrast to other methods, this DPI-ELISA screen can be performed manually or with standard laboratory automation. Furthermore, output evaluation does not require extensive computational analysis to derive a binding consensus. We could show that the DPI-ELISA screen disclosed the full spectrum of binding preferences for a given DBP. As an example, AtWRKY11 was used to demonstrate that the automated DPI-ELISA screen revealed the entire range of in vitro binding preferences. In addition, protein extracts of AtbZIP63 and the DNA-binding domain of AtWRKY33 were analyzed, which led to a refinement of their known DNA-binding consensi. Finally, we performed a DPI-ELISA screen to disclose the DNA-binding consensus of a yet uncharacterized putative DBP, AtTIFY1. A palindromic TGATCA-consensus was uncovered and we could show that the GATC-core is compulsory for AtTIFY1 binding. This specific interaction between AtTIFY1 and its DNA-binding motif was confirmed by in vivo plant one-hybrid assays in protoplasts. Thus, the value and applicability of the DPI-ELISA screen for de novo binding site identification of DBPs, also under automatized conditions, is a promising approach for a deeper understanding of gene regulation in any organism of choice.
Collapse
Affiliation(s)
- Luise H. Brand
- Plant Physiology, Center for Plant Molecular Biology, University of Tuebingen, Tuebingen, Germany
| | - Carsten Henneges
- Cognitive Systems, Center for Bioinformatics, University of Tuebingen, Tuebingen, Germany
| | - Axel Schüssler
- Cognitive Systems, Center for Bioinformatics, University of Tuebingen, Tuebingen, Germany
| | - H. Üner Kolukisaoglu
- Plant Physiology, Center for Plant Molecular Biology, University of Tuebingen, Tuebingen, Germany
- Center for Life Science Automation, Rostock, Germany
| | - Grit Koch
- Center for Life Science Automation, Rostock, Germany
| | - Niklas Wallmeroth
- Plant Physiology, Center for Plant Molecular Biology, University of Tuebingen, Tuebingen, Germany
| | - Andreas Hecker
- Plant Physiology, Center for Plant Molecular Biology, University of Tuebingen, Tuebingen, Germany
| | | | - Andreas Zell
- Cognitive Systems, Center for Bioinformatics, University of Tuebingen, Tuebingen, Germany
| | - Klaus Harter
- Plant Physiology, Center for Plant Molecular Biology, University of Tuebingen, Tuebingen, Germany
| | - Dierk Wanke
- Plant Physiology, Center for Plant Molecular Biology, University of Tuebingen, Tuebingen, Germany
- * E-mail:
| |
Collapse
|
49
|
Ben-Othman N, Courtney M, Vieira A, Pfeifer A, Druelle N, Gjernes E, Faurite B, Avolio F, Collombat P. From pancreatic islet formation to beta-cell regeneration. Diabetes Res Clin Pract 2013; 101:1-9. [PMID: 23380136 DOI: 10.1016/j.diabres.2013.01.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 01/09/2013] [Indexed: 12/15/2022]
Abstract
Diabetes mellitus represents a major healthcare burden and, due to the increasing prevalence of type I diabetes and the complications arising from current treatments, other alternative therapies must be found. Type I diabetes arises as a result of a cell-mediated autoimmune destruction of insulin producing pancreatic β-cells. Thus, a cell replacement therapy would be appropriate, using either in vitro or in vivo cell differentiation/reprogramming from different cell sources. Increasing our understanding of the molecular mechanisms controlling endocrine cell specification during pancreas morphogenesis and gaining further insight into the complex transcriptional network and signaling pathways governing β-cell development should facilitate efforts to achieve this ultimate goal, that is to regenerate insulin-producing β-cells. This review will therefore describe briefly the genetic program underlying mouse pancreas development and present new insights regarding β-cell regeneration.
Collapse
Affiliation(s)
- Nouha Ben-Othman
- Université de Nice-Sophia Antipolis, FR-06108 Nice, France; Inserm U1091, IBV, Diabetes Genetics Team, FR-06108 Nice, France; JDRF, 26 Broadway, NY-10004, USA
| | - Monica Courtney
- Université de Nice-Sophia Antipolis, FR-06108 Nice, France; Inserm U1091, IBV, Diabetes Genetics Team, FR-06108 Nice, France; JDRF, 26 Broadway, NY-10004, USA
| | - Andhira Vieira
- Université de Nice-Sophia Antipolis, FR-06108 Nice, France; Inserm U1091, IBV, Diabetes Genetics Team, FR-06108 Nice, France; JDRF, 26 Broadway, NY-10004, USA
| | - Anja Pfeifer
- Université de Nice-Sophia Antipolis, FR-06108 Nice, France; Inserm U1091, IBV, Diabetes Genetics Team, FR-06108 Nice, France; JDRF, 26 Broadway, NY-10004, USA
| | - Noémie Druelle
- Université de Nice-Sophia Antipolis, FR-06108 Nice, France; Inserm U1091, IBV, Diabetes Genetics Team, FR-06108 Nice, France; JDRF, 26 Broadway, NY-10004, USA
| | - Elisabet Gjernes
- Université de Nice-Sophia Antipolis, FR-06108 Nice, France; Inserm U1091, IBV, Diabetes Genetics Team, FR-06108 Nice, France; JDRF, 26 Broadway, NY-10004, USA
| | - Biljana Faurite
- Université de Nice-Sophia Antipolis, FR-06108 Nice, France; Inserm U1091, IBV, Diabetes Genetics Team, FR-06108 Nice, France; JDRF, 26 Broadway, NY-10004, USA
| | - Fabio Avolio
- Université de Nice-Sophia Antipolis, FR-06108 Nice, France; Inserm U1091, IBV, Diabetes Genetics Team, FR-06108 Nice, France; JDRF, 26 Broadway, NY-10004, USA
| | - Patrick Collombat
- Université de Nice-Sophia Antipolis, FR-06108 Nice, France; Inserm U1091, IBV, Diabetes Genetics Team, FR-06108 Nice, France; JDRF, 26 Broadway, NY-10004, USA.
| |
Collapse
|
50
|
Rath MF, Rohde K, Klein DC, Møller M. Homeobox genes in the rodent pineal gland: roles in development and phenotype maintenance. Neurochem Res 2013; 38:1100-12. [PMID: 23076630 PMCID: PMC3570627 DOI: 10.1007/s11064-012-0906-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 09/19/2012] [Accepted: 10/04/2012] [Indexed: 12/12/2022]
Abstract
The pineal gland is a neuroendocrine gland responsible for nocturnal synthesis of melatonin. During early development of the rodent pineal gland from the roof of the diencephalon, homeobox genes of the orthodenticle homeobox (Otx)- and paired box (Pax)-families are expressed and are essential for normal pineal development consistent with the well-established role that homeobox genes play in developmental processes. However, the pineal gland appears to be unusual because strong homeobox gene expression persists in the pineal gland of the adult brain. Accordingly, in addition to developmental functions, homeobox genes appear to be key regulators in postnatal phenotype maintenance in this tissue. In this paper, we review ontogenetic and phylogenetic aspects of pineal development and recent progress in understanding the involvement of homebox genes in rodent pineal development and adult function. A working model is proposed for understanding the sequential action of homeobox genes in controlling development and mature circadian function of the mammalian pinealocyte based on knowledge from detailed developmental and daily gene expression analyses in rats, the pineal phenotypes of homebox gene-deficient mice and studies on development of the retinal photoreceptor; the pinealocyte and retinal photoreceptor share features not seen in other tissues and are likely to have evolved from the same ancestral photodetector cell.
Collapse
Affiliation(s)
- Martin F Rath
- Department of Neuroscience and Pharmacology, Panum Institute 24.2, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen 2200, Denmark.
| | | | | | | |
Collapse
|