1
|
Hill DJ, Hill TG. Maternal diet during pregnancy and adaptive changes in the maternal and fetal pancreas have implications for future metabolic health. Front Endocrinol (Lausanne) 2024; 15:1456629. [PMID: 39377073 PMCID: PMC11456468 DOI: 10.3389/fendo.2024.1456629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/28/2024] [Indexed: 10/09/2024] Open
Abstract
Fetal and neonatal development is a critical period for the establishment of the future metabolic health and disease risk of an individual. Both maternal undernutrition and overnutrition can result in abnormal fetal organ development resulting in inappropriate birth size, child and adult obesity, and increased risk of Type 2 diabetes and cardiovascular diseases. Inappropriate adaptive changes to the maternal pancreas, placental function, and the development of the fetal pancreas in response to nutritional stress during pregnancy are major contributors to a risk trajectory in the offspring. This interconnected maternal-placental-fetal metabolic axis is driven by endocrine signals in response to the availability of nutritional metabolites and can result in cellular stress and premature aging in fetal tissues and the inappropriate expression of key genes involved in metabolic control as a result of long-lasting epigenetic changes. Such changes result is insufficient pancreatic beta-cell mass and function, reduced insulin sensitivity in target tissues such as liver and white adipose and altered development of hypothalamic satiety centres and in basal glucocorticoid levels. Whilst interventions in the obese mother such as dieting and increased exercise, or treatment with insulin or metformin in mothers who develop gestational diabetes, can improve metabolic control and reduce the risk of a large-for-gestational age infant, their effectiveness in changing the adverse metabolic trajectory in the child is as yet unclear.
Collapse
Affiliation(s)
- David J. Hill
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON, Canada
- Departments of Medicine, Physiology and Pharmacology, Western University, London, ON, Canada
| | - Thomas G. Hill
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
2
|
Mathisen AF, Larsen U, Kavli N, Unger L, Daian LM, Vacaru AM, Vacaru AM, Herrera PL, Ghila L, Chera S. Moderate beta-cell ablation triggers synergic compensatory mechanisms even in the absence of overt metabolic disruption. Commun Biol 2024; 7:833. [PMID: 38982170 PMCID: PMC11233560 DOI: 10.1038/s42003-024-06527-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024] Open
Abstract
Regeneration, the ability to replace injured tissues and organs, is a phenomenon commonly associated with lower vertebrates but is also observed in mammals, in specific tissues. In this study, we investigated the regenerative potential of pancreatic islets following moderate beta-cell loss in mice. Using a rapid model of moderate ablation, we observed a compensatory response characterized by transient inflammation and proliferation signatures, ultimately leading to the recovery of beta-cell identity and function. Interestingly, this proliferative response occurred independently of inflammation, as demonstrated in ablated immunodeficient mice. Furthermore, exposure to high-fat diet stimulated beta-cell proliferation but negatively impacted beta-cell function. In contrast, an equivalent slower ablation model revealed a delayed but similar proliferative response, suggesting proliferation as a common regenerative response. However, high-fat diet failed to promote proliferation in this model, indicating a differential response to metabolic stressors. Overall, our findings shed light on the complex interplay between beta-cell loss, inflammation, and stress in modulating pancreatic islet regeneration. Understanding these mechanisms could pave the way for novel therapeutic strategies based on beta-cell proliferation.
Collapse
Affiliation(s)
- Andreas Frøslev Mathisen
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ulrik Larsen
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Natalie Kavli
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lucas Unger
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Laura Maria Daian
- BetaUpreg Research Group, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Andrei Mircea Vacaru
- BetaUpreg Research Group, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Ana-Maria Vacaru
- BetaUpreg Research Group, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Pedro Luis Herrera
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Luiza Ghila
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Simona Chera
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
3
|
Gu G, Brown M, Agan V, Nevills S, Hu R, Simmons A, Xu Y, Yang Y, Yagan M, Najam S, Dadi P, Sampson L, Magnuson M, Jacobson D, Lau K, Hodges E. Endocrine islet β-cell subtypes with differential function are derived from biochemically distinct embryonic endocrine islet progenitors that are regulated by maternal nutrients. RESEARCH SQUARE 2024:rs.3.rs-3946483. [PMID: 38496675 PMCID: PMC10942487 DOI: 10.21203/rs.3.rs-3946483/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Endocrine islet b cells comprise heterogenous cell subsets. Yet when/how these subsets are produced and how stable they are remain unknown. Addressing these questions is important for preventing/curing diabetes, because lower numbers of b cells with better secretory function is a high risk of this disease. Using combinatorial cell lineage tracing, scRNA-seq, and DNA methylation analysis, we show here that embryonic islet progenitors with distinct gene expression and DNA methylation produce b-cell subtypes of different function and viability in adult mice. The subtype with better function is enriched for genes involved in vesicular production/trafficking, stress response, and Ca2+-secretion coupling, which further correspond to differential DNA methylation in putative enhancers of these genes. Maternal overnutrition, a major diabetes risk factor, reduces the proportion of endocrine progenitors of the b-cell subtype with better-function via deregulating DNA methyl transferase 3a. Intriguingly, the gene signature that defines mouse b-cell subtypes can reliably divide human cells into two sub-populations while the proportion of b cells with better-function is reduced in diabetic donors. The implication of these results is that modulating DNA methylation in islet progenitors using maternal food supplements can be explored to improve b-cell function in the prevention and therapy of diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yilin Yang
- Vanderbilty University School of Medicine
| | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Onodera T, Kim DS, Ye R, Wang MY, Chen S, Field BC, Straub L, Sun XN, Li C, Lee C, Paredes M, Crewe C, Zhao S, Kusminski CM, Gordillo R, Scherer PE. Protective roles of adiponectin and molecular signatures of HNF4α and PPARα as downstream targets of adiponectin in pancreatic β cells. Mol Metab 2023; 78:101821. [PMID: 37806486 PMCID: PMC10598053 DOI: 10.1016/j.molmet.2023.101821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023] Open
Abstract
The disease progression of the metabolic syndrome is associated with prolonged hyperlipidemia and insulin resistance, eventually giving rise to impaired insulin secretion, often concomitant with hypoadiponectinemia. As an adipose tissue derived hormone, adiponectin is beneficial for insulin secretion and β cell health and differentiation. However, the down-stream pathway of adiponectin in the pancreatic islets has not been studied extensively. Here, along with the overall reduction of endocrine pancreatic function in islets from adiponectin KO mice, we examine PPARα and HNF4α as additional down-regulated transcription factors during a prolonged metabolic challenge. To elucidate the function of β cell-specific PPARα and HNF4α expression, we developed doxycycline inducible pancreatic β cell-specific PPARα (β-PPARα) and HNF4α (β-HNF4α) overexpression mice. β-PPARα mice exhibited improved protection from lipotoxicity, but elevated β-oxidative damage in the islets, and also displayed lowered phospholipid levels and impaired glucose-stimulated insulin secretion. β-HNF4α mice showed a more severe phenotype when compared to β-PPARα mice, characterized by lower body weight, small islet mass and impaired insulin secretion. RNA-sequencing of the islets of these models highlights overlapping yet unique roles of β-PPARα and β-HNF4α. Given that β-HNF4α potently induces PPARα expression, we define a novel adiponectin-HNF4α-PPARα cascade. We further analyzed downstream genes consistently regulated by this axis. Among them, the islet amyloid polypeptide (IAPP) gene is an important target and accumulates in adiponectin KO mice. We propose a new mechanism of IAPP aggregation in type 2 diabetes through reduced adiponectin action.
Collapse
Affiliation(s)
- Toshiharu Onodera
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Dae-Seok Kim
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Risheng Ye
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - May-Yun Wang
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Shiuhwei Chen
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Bianca C Field
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Leon Straub
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Xue-Nan Sun
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Chao Li
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Charlotte Lee
- Center for Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Megan Paredes
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Clair Crewe
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Shangang Zhao
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Christine M Kusminski
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Ruth Gordillo
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, United States.
| |
Collapse
|
5
|
Rastinejad F. The protein architecture and allosteric landscape of HNF4α. Front Endocrinol (Lausanne) 2023; 14:1219092. [PMID: 37732120 PMCID: PMC10507258 DOI: 10.3389/fendo.2023.1219092] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/01/2023] [Indexed: 09/22/2023] Open
Abstract
Hepatocyte nuclear factor 4 alpha (HNF4α) is a multi-faceted nuclear receptor responsible for governing the development and proper functioning of liver and pancreatic islet cells. Its transcriptional functions encompass the regulation of vital metabolic processes including cholesterol and fatty acid metabolism, and glucose sensing and control. Various genetic mutations and alterations in HNF4α are associated with diabetes, metabolic disorders, and cancers. From a structural perspective, HNF4α is one of the most comprehensively understood nuclear receptors due to its crystallographically observed architecture revealing interconnected DNA binding domains (DBDs) and ligand binding domains (LBDs). This review discusses key properties of HNF4α, including its mode of homodimerization, its binding to fatty acid ligands, the importance of post-translational modifications, and the mechanistic basis for allosteric functions. The surfaces linking HNF4α's DBDs and LBDs create a convergence zone that allows signals originating from any one domain to influence distant domains. The HNF4α-DNA complex serves as a prime illustration of how nuclear receptors utilize individual domains for specific functions, while also integrating these domains to create cohesive higher-order architectures that allow signal responsive functions.
Collapse
Affiliation(s)
- Fraydoon Rastinejad
- Nuffield Department of Medicine, Target Discovery Institute (NDMRB), University of Oxford, Oxford, United Kingdom
| |
Collapse
|
6
|
Peng G, Mosleh E, Yuhas A, Katada K, Cherry C, Golson ML. FOXM1 acts sexually dimorphically to regulate functional β-cell mass. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523673. [PMID: 36711451 PMCID: PMC9882186 DOI: 10.1101/2023.01.12.523673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The transcription factor FOXM1 regulates β-cell proliferation and insulin secretion. Our previous work demonstrates that expressing an activated form of FOXM1 (FOXM1*) in β cells increases β-cell proliferation and mass in aged male mice. Additionally, FOXM1* enhances β-cell function even in young mice, in which no β-cell mass elevation occurs. Here, we demonstrate that FOXM1 acts in a sexually dimorphic manner in the β cell. Expression of FOXM1* in female mouse β cells does not affect β-cell proliferation or glucose tolerance. Transduction of male but not female human islets with FOXM1* enhances insulin secretion in response to elevated glucose. Estrogen contributes to diabetes susceptibility differences between males and females, and the estrogen receptor (ER)α is the primary mediator of β-cell estrogen signaling. We show that FOXM1* can rescue impaired glucose tolerance in female mice with a pancreas-wide ERα deletion. Further, FOXM1 and ERα binding sites overlap with each other and with other β-cell-enriched transcription factors, including ISL1, PAX6, MAF, and GATA. These data indicate that FOMX1 and ERα cooperate to regulate β-cell function and suggest a general mechanism contributing to the lower incidence of diabetes observed in women.
Collapse
|
7
|
Jiang Y, Chen A, Kline D, Liu Q, Ma J, Wang Y, Zhang T, Qian J, Nelson L, Prasadan K, Hu B, Gittes GK, Xiao X. Polarized macrophages promote gestational beta cell growth through extracellular signal-regulated kinase 5 signalling. Diabetes Obes Metab 2022; 24:1721-1733. [PMID: 35546452 DOI: 10.1111/dom.14744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/24/2022] [Accepted: 04/29/2022] [Indexed: 12/25/2022]
Abstract
AIM To show that depletion of pancreatic macrophages impairs gestational beta cell proliferation and leads to glucose intolerance. MATERIALS AND METHODS Genetic animal models were applied to study the effects of depletion of pancreatic macrophges on gestational beta-cell proliferaiton and glucose response. The crosstalk between macrophages and beta-cells was studied in vivo using beta-cell-specific extracellular-signal-regulated kinase 5 (ERK5) knockout and epidermal growth receptor (EGFR) knockout mice, and in vitro using a co-culture system. RESULTS Beta cell-derived placental growth factor (PlGF) recruited naïve macrophages and polarized them towards an M2-like phenotype. These macrophages then secreted epidermal growth factor (EGF), which activated extracellular signal-regulated kinase 5 (ERK5) signalling in beta cells to promote gestational beta cell proliferation. On the other hand, activation of ERK5 signalling in beta cells likely, in turn, enhanced the production and secretion of PlGF by beta cells. CONCLUSIONS Our study shows a regulatory loop between macrophages and beta cells through PlGF/EGF/ERK5 signalling cascades to regulate gestational beta cell growth.
Collapse
Affiliation(s)
- Yinan Jiang
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Apeng Chen
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Diana Kline
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Qun Liu
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jie Ma
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yan Wang
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ting Zhang
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jieqi Qian
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Laura Nelson
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Krishna Prasadan
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Baoli Hu
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - George K Gittes
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xiangwei Xiao
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
8
|
Eguchi N, Toribio AJ, Alexander M, Xu I, Whaley DL, Hernandez LF, Dafoe D, Ichii H. Dysregulation of β-Cell Proliferation in Diabetes: Possibilities of Combination Therapy in the Development of a Comprehensive Treatment. Biomedicines 2022; 10:biomedicines10020472. [PMID: 35203680 PMCID: PMC8962301 DOI: 10.3390/biomedicines10020472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 02/01/2023] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder characterized by chronic hyperglycemia as a result of insufficient insulin levels and/or impaired function as a result of autoimmune destruction or insulin resistance. While Type 1 DM (T1DM) and Type 2 DM (T2DM) occur through different pathological processes, both result in β-cell destruction and/or dysfunction, which ultimately lead to insufficient β-cell mass to maintain normoglycemia. Therefore, therapeutic agents capable of inducing β-cell proliferation is crucial in treating and reversing diabetes; unfortunately, adult human β-cell proliferation has been shown to be very limited (~0.2% of β-cells/24 h) and poorly responsive to many mitogens. Furthermore, diabetogenic insults result in damage to β cells, making it ever more difficult to induce proliferation. In this review, we discuss β-cell mass/proliferation pathways dysregulated in diabetes and current therapeutic agents studied to induce β-cell proliferation. Furthermore, we discuss possible combination therapies of proliferation agents with immunosuppressants and antioxidative therapy to improve overall long-term outcomes of diabetes.
Collapse
|
9
|
Barth R, Ruoso C, Ferreira SM, de Ramos FC, Lima FB, Boschero AC, Santos GJD. Hepatocyte Nuclear Factor 4-α (HNF4α) controls the insulin resistance-induced pancreatic β-cell mass expansion. Life Sci 2022; 289:120213. [PMID: 34902439 DOI: 10.1016/j.lfs.2021.120213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/17/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Regardless of the etiology, any type of DM presents a reduction of insulin-secreting cell mass, so it is important to investigate pathways that induce the increase of this cell mass. AIM Based on the fact that (1) HNF4α is crucial for β-cell proliferation, (2) DEX-induced IR promotes β-cell mass expansion, and (3) the stimulation of β-cell mass expansion may be an important target for DM therapies, we aimed to investigate whether DEX-induced proliferation of β pancreatic cells is dependent on HNF4α. METHODS We used WildType (WT) and Knockout (KO) mice for HNF4-α, treated or not with 100 mg/Kg/day of DEX, for 5 consecutive days. One day after the last injection of DEX the IR was confirmed by ipITT and the mice were euthanized for pancreas removal. RESULTS In comparison to WT, KO mice presented increased glucose tolerance, lower fasting glucose and increased glucose-stimulates insulin secretion (GSIS). DEX induced IR in both KO and WT mice. In addition, DEX-induced β-cell mass expansion and an increase in the Ki67 immunostaining were observed only in WT mice, evidencing that IR-induced β-cell mass expansion is dependent on HNF4α. Also, we observed that DEX-treatment, in an HNF4α-dependent way, promoted an increase in PDX1, PAX4 and NGN3 gene expression. CONCLUSIONS Our results strongly suggest that DEX-induced IR promotes β-cell mass expansion through processes of proliferation and neogenesis that depend on the HNF4α activity, pointing to HNF4α as a possible therapeutic target in DM treatment.
Collapse
Affiliation(s)
- Robson Barth
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil
| | - Carolina Ruoso
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil
| | - Sandra Mara Ferreira
- Laboratory of endocrine pancreas and metabolism - LAPEM, Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, 13083-862 Campinas, Brazil
| | - Francieli Caroline de Ramos
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil
| | - Fernanda Barbosa Lima
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil
| | - Antônio Carlos Boschero
- Laboratory of endocrine pancreas and metabolism - LAPEM, Department of Structural and Functional Biology, Institute of Biology, State University of Campinas - UNICAMP, 13083-862 Campinas, Brazil
| | - Gustavo Jorge Dos Santos
- Islet Biology and Metabolism Lab - I.B.M. Lab, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis 88040-900, Santa Catarina, Brazil; Multicenter Graduate Program in Physiological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, Brazil.
| |
Collapse
|
10
|
Ntunzwenimana JC, Boucher G, Paquette J, Gosselin H, Alikashani A, Morin N, Beauchamp C, Thauvette L, Rivard MÈ, Dupuis F, Deschênes S, Foisy S, Latour F, Lavallée G, Daly MJ, Xavier RJ, Charron G, Goyette P, Rioux JD. Functional screen of inflammatory bowel disease genes reveals key epithelial functions. Genome Med 2021; 13:181. [PMID: 34758847 PMCID: PMC8582123 DOI: 10.1186/s13073-021-00996-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/21/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Genetic studies have been tremendously successful in identifying genomic regions associated with a wide variety of phenotypes, although the success of these studies in identifying causal genes, their variants, and their functional impacts has been more limited. METHODS We identified 145 genes from IBD-associated genomic loci having endogenous expression within the intestinal epithelial cell compartment. We evaluated the impact of lentiviral transfer of the open reading frame (ORF) of these IBD genes into the HT-29 intestinal epithelial cell line via transcriptomic analyses. By comparing the genes in which expression was modulated by each ORF, as well as the functions enriched within these gene lists, we identified ORFs with shared impacts and their putative disease-relevant biological functions. RESULTS Analysis of the transcriptomic data for cell lines expressing the ORFs for known causal genes such as HNF4a, IFIH1, and SMAD3 identified functions consistent with what is already known for these genes. These analyses also identified two major clusters of genes: Cluster 1 contained the known IBD causal genes IFIH1, SBNO2, NFKB1, and NOD2, as well as genes from other IBD loci (ZFP36L1, IRF1, GIGYF1, OTUD3, AIRE and PITX1), whereas Cluster 2 contained the known causal gene KSR1 and implicated DUSP16 from another IBD locus. Our analyses highlight how multiple IBD gene candidates can impact on epithelial structure and function, including the protection of the mucosa from intestinal microbiota, and demonstrate that DUSP16 acts a regulator of MAPK activity and contributes to mucosal defense, in part via its regulation of the polymeric immunoglobulin receptor, involved in the protection of the intestinal mucosa from enteric microbiota. CONCLUSIONS This functional screen, based on expressing IBD genes within an appropriate cellular context, in this instance intestinal epithelial cells, resulted in changes to the cell's transcriptome that are relevant to their endogenous biological function(s). This not only helped in identifying likely causal genes within genetic loci but also provided insight into their biological functions. Furthermore, this work has highlighted the central role of intestinal epithelial cells in IBD pathophysiology, providing a scientific rationale for a drug development strategy that targets epithelial functions in addition to the current therapies targeting immune functions.
Collapse
Affiliation(s)
- Jessy Carol Ntunzwenimana
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Gabrielle Boucher
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Jean Paquette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Hugues Gosselin
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Azadeh Alikashani
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Nicolas Morin
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Claudine Beauchamp
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Louise Thauvette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Marie-Ève Rivard
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Frédérique Dupuis
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Sonia Deschênes
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Sylvain Foisy
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Frédéric Latour
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Geneviève Lavallée
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Mark J Daly
- Massachusetts General Hospital, Boston, MA, USA
- The Broad Institute, Cambridge, MA, USA
| | - Ramnik J Xavier
- Massachusetts General Hospital, Boston, MA, USA
- The Broad Institute, Cambridge, MA, USA
| | - Guy Charron
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Philippe Goyette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - John D Rioux
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada.
- Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
11
|
Altman MK, Schaub CM, Dadi PK, Dickerson MT, Zaborska KE, Nakhe AY, Graff SM, Galletta TJ, Amarnath G, Thorson AS, Gu G, Jacobson DA. TRPM7 is a crucial regulator of pancreatic endocrine development and high-fat-diet-induced β-cell proliferation. Development 2021; 148:dev194928. [PMID: 34345920 PMCID: PMC8406533 DOI: 10.1242/dev.194928] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 07/20/2021] [Indexed: 12/31/2022]
Abstract
The melastatin subfamily of the transient receptor potential channels (TRPM) are regulators of pancreatic β-cell function. TRPM7 is the most abundant islet TRPM channel; however, the role of TRPM7 in β-cell function has not been determined. Here, we used various spatiotemporal transgenic mouse models to investigate how TRPM7 knockout influences pancreatic endocrine development, proliferation and function. Ablation of TRPM7 within pancreatic progenitors reduced pancreatic size, and α-cell and β-cell mass. This resulted in modestly impaired glucose tolerance. However, TRPM7 ablation following endocrine specification or in adult mice did not impact endocrine expansion or glucose tolerance. As TRPM7 regulates cell proliferation, we assessed how TRPM7 influences β-cell hyperplasia under insulin-resistant conditions. β-Cell proliferation induced by high-fat diet was significantly decreased in TRPM7-deficient β-cells. The endocrine roles of TRPM7 may be influenced by cation flux through the channel, and indeed we found that TRPM7 ablation altered β-cell Mg2+ and reduced the magnitude of elevation in β-cell Mg2+ during proliferation. Together, these findings revealed that TRPM7 controls pancreatic development and β-cell proliferation, which is likely due to regulation of Mg2+ homeostasis.
Collapse
Affiliation(s)
- Molly K. Altman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Charles M. Schaub
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Prasanna K. Dadi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Matthew T. Dickerson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Karolina E. Zaborska
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Arya Y. Nakhe
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Sarah M. Graff
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Thomas J. Galletta
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Gautami Amarnath
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
- Molecular Neurophysiology, Institute of Applied Physiology, University of Ulm, 89081 Ulm, Germany
| | - Ariel S. Thorson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Guoqiang Gu
- Vanderbilt Program in Developmental Biology, Vanderbilt Center for Stem Cell Biology, Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - David A. Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| |
Collapse
|
12
|
Cheng J, Li M, Tzeng CM, Gou X, Chen S. Suppression of Tumorigenicity 5 Ameliorates Tumor Characteristics of Invasive Breast Cancer Cells via ERK/JNK Pathway. Front Oncol 2021; 11:621500. [PMID: 34395234 PMCID: PMC8356645 DOI: 10.3389/fonc.2021.621500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 06/04/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Suppression of tumorigenicity 5 (ST5) has been considered as a tumor suppressor gene in HeLa tumor cells. However, its role in the progression of breast cancer remains vague. METHODS Online database analysis was determined by Oncomine and Breast Cancer Gene-Expression Miner v4.4 (bc-GenExMiner v4.4). Tumor biology behaviors were measured by MTT assay, wound healing model, Transwell and Flow cytometry assays. Methylation-specific PCR (MSP) was employed to detect promoter methylation. RESULTS Low level of ST5 was observed in breast cancer specimens, particularly in recurrent, invasive breast cancer cases compared to para-carcinoma tissue or non-invasive breast cancer. The downregulation of ST5 was also proved in MDA-MB-231 and SKBR3 cell lines with a high invasive capability as compared to MCF-7 cell with a low invasive capability. ST5 was negatively associated with pathological stages of breast cancer. ST5-downregulation promoted, while ST5-upregulation inhibited the progression of cell proliferation, cell cycle and migration of MDA-MB-231 cells. Additionally, ST5 knockdown inhibited, whereas ST5 overexpression promoted apoptosis of MDA-MB-231 cells. However, ST5 modification, either upregulation or downregulation, had no significant impact on tumor behaviors of MCF-7 cells. Mechanistically, ST5 protein ablation activated, while ST5-upregulation repressed the activities of phosphorylated ERK1/2 and JNK, and subsequently the expression of c-Myc. PD98059-mediated ERK1/2 inhibition abolished the stimulatory effects of ST5-depletion on ERK1/2/JNK/c-Myc signaling axis, and ST5 depletion-mediated cell over-proliferation and migration. Of note, ST5 reduction in invasive breast cancer cells should implicate in the hypermethylation of ST5 promoter region. CONCLUSION Our findings suggest that ST5 potentially acts as a tumor suppressor gene in invasive breast cancer through regulating ERK/JNK signaling pathway and provide a novel insight for breast cancer treatment.
Collapse
Affiliation(s)
- Jianghong Cheng
- Shaanxi Key Laboratory of Brain Disorders and School of Basic Medical Science, Xi’an Medical University, Xi’an, China
| | - Mingli Li
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, China
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Otolaryngology Head and Neck Surgery, Xiamen, China
| | - Chi-Meng Tzeng
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Science, Xiamen University, Xiamen, China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders and School of Basic Medical Science, Xi’an Medical University, Xi’an, China
- Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
- Academician Workstation of Chen Zhi-nan, Xi’an Medical University, Xi’an, China
| | - Shuai Chen
- Shaanxi Key Laboratory of Brain Disorders and School of Basic Medical Science, Xi’an Medical University, Xi’an, China
- Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
- Academician Workstation of Chen Zhi-nan, Xi’an Medical University, Xi’an, China
| |
Collapse
|
13
|
Szlapinski SK, Hill DJ. Metabolic Adaptations to Pregnancy in Healthy and Gestational Diabetic Pregnancies: The Pancreas - Placenta Axis. Curr Vasc Pharmacol 2021; 19:141-153. [PMID: 32196450 DOI: 10.2174/1570161118666200320111209] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/20/2020] [Accepted: 03/01/2020] [Indexed: 12/16/2022]
Abstract
Normal pregnancy is associated with increased insulin resistance as a metabolic adaptation to the nutritional demands of the placenta and fetus, and this is amplified in obese mothers. Insulin resistance is normally compensated for by an adaptive increase in pancreatic β-cell mass together with enhanced glucose-stimulated insulin release. Placentally-derived hormones and growth factors are central to the altered pancreatic morphology and function. A failure of β-cells to undergo adaptive change after the first trimester has been linked with gestational diabetes. In the pregnant mouse, an increase in β-cell replication contributes to a 2-3-fold increase in mass peaking in late gestation, depending on the proliferation of existing β-cells, the differentiation of resident progenitor β-cells, or islet cell transdifferentiation. Using mouse models and human studies placenta- and islet of Langerhans-derived molecules have been identified that are likely to contribute to the metabolic adaptations to pregnancy and whose physiology is altered in the obese, glucose-intolerant mother. Maternal obesity during pregnancy can create a pro-inflammatory environment that can disrupt the response of the β-cells to the endocrine signals of pregnancy and limit the adaptive changes in β-cell mass and function, resulting in an increased risk of gestational diabetes.
Collapse
Affiliation(s)
- Sandra K Szlapinski
- Lawson Health Research Institute, St. Joseph's Health Care, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| | - David J Hill
- Lawson Health Research Institute, St. Joseph's Health Care, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
| |
Collapse
|
14
|
A Brief Review of the Mechanisms of β-Cell Dedifferentiation in Type 2 Diabetes. Nutrients 2021; 13:nu13051593. [PMID: 34068827 PMCID: PMC8151793 DOI: 10.3390/nu13051593] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 01/09/2023] Open
Abstract
Diabetes is a metabolic disease characterized by hyperglycemia. Over 90% of patients with diabetes have type 2 diabetes. Pancreatic β-cells are endocrine cells that produce and secrete insulin, an essential endocrine hormone that regulates blood glucose levels. Deficits in β-cell function and mass play key roles in the onset and progression of type 2 diabetes. Apoptosis has been considered as the main contributor of β-cell dysfunction and decrease in β-cell mass for a long time. However, recent studies suggest that β-cell failure occurs mainly due to increased β-cell dedifferentiation rather than limited β-cell proliferation or increased β-cell death. In this review, we summarize the current advances in the understanding of the pancreatic β-cell dedifferentiation process including potential mechanisms. A better understanding of β-cell dedifferentiation process will help to identify novel therapeutic targets to prevent and/or reverse β-cell loss in type 2 diabetes.
Collapse
|
15
|
Ornoy A, Becker M, Weinstein-Fudim L, Ergaz Z. Diabetes during Pregnancy: A Maternal Disease Complicating the Course of Pregnancy with Long-Term Deleterious Effects on the Offspring. A Clinical Review. Int J Mol Sci 2021; 22:2965. [PMID: 33803995 PMCID: PMC7999044 DOI: 10.3390/ijms22062965] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 02/28/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
In spite of the huge progress in the treatment of diabetes mellitus, we are still in the situation that both pregestational (PGDM) and gestational diabetes (GDM) impose an additional risk to the embryo, fetus, and course of pregnancy. PGDM may increase the rate of congenital malformations, especially cardiac, nervous system, musculoskeletal system, and limbs. PGDM may interfere with fetal growth, often causing macrosomia, but in the presence of severe maternal complications, especially nephropathy, it may inhibit fetal growth. PGDM may also induce a variety of perinatal complications such as stillbirth and perinatal death, cardiomyopathy, respiratory morbidity, and perinatal asphyxia. GDM that generally develops in the second half of pregnancy induces similar but generally less severe complications. Their severity is higher with earlier onset of GDM and inversely correlated with the degree of glycemic control. Early initiation of GDM might even cause some increase in the rate of congenital malformations. Both PGDM and GDM may cause various motor and behavioral neurodevelopmental problems, including an increased incidence of attention deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). Most complications are reduced in incidence and severity with the improvement in diabetic control. Mechanisms of diabetic-induced damage in pregnancy are related to maternal and fetal hyperglycemia, enhanced oxidative stress, epigenetic changes, and other, less defined, pathogenic mechanisms.
Collapse
Affiliation(s)
- Asher Ornoy
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
- Laboratory of Teratology, Department of Medical Neurobiology, Hebrew University Hadassah Medical School, Jerusalem 91120, Israel; (L.W.-F.); (Z.E.)
| | - Maria Becker
- Adelson School of Medicine, Ariel University, Ariel 40700, Israel;
| | - Liza Weinstein-Fudim
- Laboratory of Teratology, Department of Medical Neurobiology, Hebrew University Hadassah Medical School, Jerusalem 91120, Israel; (L.W.-F.); (Z.E.)
| | - Zivanit Ergaz
- Laboratory of Teratology, Department of Medical Neurobiology, Hebrew University Hadassah Medical School, Jerusalem 91120, Israel; (L.W.-F.); (Z.E.)
- Medical Center, Hadassah Hebrew University, Mount Scopus, Jerusalem 91240, Israel
| |
Collapse
|
16
|
Salazar-Petres ER, Sferruzzi-Perri AN. Pregnancy-induced changes in β-cell function: what are the key players? J Physiol 2021; 600:1089-1117. [PMID: 33704799 DOI: 10.1113/jp281082] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/17/2021] [Indexed: 12/11/2022] Open
Abstract
Maternal metabolic adaptations during pregnancy ensure appropriate nutrient supply to the developing fetus. This is facilitated by reductions in maternal peripheral insulin sensitivity, which enables glucose to be available in the maternal circulation for transfer to the fetus for growth. To balance this process and avoid excessive hyperglycaemia and glucose intolerance in the mother during pregnancy, maternal pancreatic β-cells undergo remarkable changes in their function including increasing their proliferation and glucose-stimulated insulin secretion. In this review we examine how placental and maternal hormones work cooperatively to activate several signalling pathways, transcription factors and epigenetic regulators to drive adaptations in β-cell function during pregnancy. We also explore how adverse maternal environmental conditions, including malnutrition, obesity, circadian rhythm disruption and environmental pollutants, may impact the endocrine and molecular mechanisms controlling β-cell adaptations during pregnancy. The available data from human and experimental animal studies highlight the need to better understand how maternal β-cells integrate the various environmental, metabolic and endocrine cues and thereby determine appropriate β-cell adaptation during gestation. In doing so, these studies may identify targetable pathways that could be used to prevent not only the development of pregnancy complications like gestational diabetes that impact maternal and fetal wellbeing, but also more generally the pathogenesis of other metabolic conditions like type 2 diabetes.
Collapse
Affiliation(s)
- Esteban Roberto Salazar-Petres
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Amanda Nancy Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| |
Collapse
|
17
|
Moin ASM, Zeng K, Rizza RA, Dhawan S, Butler AE. Chromogranin A-positive hormone-negative endocrine cells in pancreas in human pregnancy. ENDOCRINOLOGY DIABETES & METABOLISM 2021; 4:e00223. [PMID: 33855223 PMCID: PMC8029563 DOI: 10.1002/edm2.223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 11/22/2022]
Abstract
Introduction We sought to determine whether chromogranin A‐positive hormone‐negative (CPHN) endocrine cells are increased in the pancreas of pregnant women, offering potential evidence in support of neogenesis. Methods Autopsy pancreata from pregnant women (n = 14) and age‐matched non‐pregnant control women (n = 9) were obtained. Staining of pancreatic sections for chromogranin A, insulin and a cocktail of glucagon, somatostatin, pancreatic polypeptide and ghrelin was undertaken, with subsequent evaluation for CPHN cell frequency. Results The frequency of clustered β‐cells was increased in pregnant compared to non‐pregnant subjects (46.6 ± 5.0 vs. 31.8 ± 5.0% clustered β‐cells of total clustered endocrine cells, pregnant vs. non‐pregnant, p < .05). Frequency of endocrine cocktail cells was lower in pregnant women than non‐pregnant women (36.2 ± 4.0 vs. 57.0 ± 6.8% clustered endocrine cocktail cells of total clustered endocrine cells, pregnant vs. non‐pregnant, p < .01). No difference in frequency of CPHN cells was found in islets, nor in clustered or single cells scattered throughout the exocrine pancreas, between pregnant and non‐pregnant women. The frequency of CPHN cells in pregnancy was independent of the number of pregnancies (gravidity). Conclusions Our findings of no increase in CPHN cell frequency in pancreas of pregnant women suggest that this potential β‐cell regenerative mechanism is not that by which the increased β‐cell mass of pregnancy is achieved. However, an increase in the percentage of clustered β‐cells was found in pregnancy, with decreased frequency of other endocrine cells in clusters, suggesting a compensatory shift from other pancreatic endocrine cell types to β‐cells as a mechanism to meet the increased insulin demands of pregnancy.
Collapse
Affiliation(s)
- Abu Saleh Md Moin
- Diabetes Research Center (DRC) Qatar Biomedical Research Institute (QBRI) Hamad Bin Khalifa University (HBKU) Qatar Foundation (QF) Doha Qatar
| | - Kylie Zeng
- Larry L. Hillblom Islet Research Center David Geffen School of Medicine University of California Los Angeles Los Angeles CA USA
| | - Robert A Rizza
- Division of Endocrinology Diabetes, Metabolism, and Nutrition Mayo Clinic College of Medicine Rochester MN USA
| | - Sangeeta Dhawan
- Diabetes and Metabolism Research Institute City of Hope Duarte CA USA
| | - Alexandra E Butler
- Diabetes Research Center (DRC) Qatar Biomedical Research Institute (QBRI) Hamad Bin Khalifa University (HBKU) Qatar Foundation (QF) Doha Qatar
| |
Collapse
|
18
|
Meijer I, Willems S, Ni X, Heering J, Chaikuad A, Merk D. Chemical Starting Matter for HNF4α Ligand Discovery and Chemogenomics. Int J Mol Sci 2020; 21:E7895. [PMID: 33114319 PMCID: PMC7660650 DOI: 10.3390/ijms21217895] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/16/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Hepatocyte nuclear factor 4α (HNF4α) is a ligand-sensing transcription factor and presents as a potential drug target in metabolic diseases and cancer. In humans, mutations in the HNF4α gene cause maturity-onset diabetes of the young (MODY), and the elevated activity of this protein has been associated with gastrointestinal cancers. Despite the high therapeutic potential, available ligands and structure-activity relationship knowledge for this nuclear receptor are scarce. Here, we disclose a chemically diverse collection of orthogonally validated fragment-like activators as well as inverse agonists, which modulate HNF4α activity in a low micromolar range. These compounds demonstrate the druggability of HNF4α and thus provide a starting point for medicinal chemistry as well as an early tool for chemogenomics.
Collapse
Affiliation(s)
- Isabelle Meijer
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany; (I.M.); (S.W.); (X.N.); (A.C.)
| | - Sabine Willems
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany; (I.M.); (S.W.); (X.N.); (A.C.)
| | - Xiaomin Ni
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany; (I.M.); (S.W.); (X.N.); (A.C.)
- Structural Genomics Consortium, BMLS, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt, Germany
| | - Jan Heering
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany;
| | - Apirat Chaikuad
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany; (I.M.); (S.W.); (X.N.); (A.C.)
- Structural Genomics Consortium, BMLS, Goethe-University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany; (I.M.); (S.W.); (X.N.); (A.C.)
| |
Collapse
|
19
|
MafB Is Important for Pancreatic β-Cell Maintenance under a MafA-Deficient Condition. Mol Cell Biol 2019; 39:MCB.00080-19. [PMID: 31208980 PMCID: PMC6692125 DOI: 10.1128/mcb.00080-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/28/2019] [Indexed: 12/28/2022] Open
Abstract
The pancreatic-islet-enriched transcription factors MafA and MafB have unique expression patterns in β cells in rodents. MafA is specifically expressed in β cells and is a key regulatory factor for maintaining adult β-cell function, whereas MafB plays an essential role in β-cell development during embryogenesis, and its expression in β cells gradually decreases and is restricted to α cells after birth in rodents. The pancreatic-islet-enriched transcription factors MafA and MafB have unique expression patterns in β cells in rodents. MafA is specifically expressed in β cells and is a key regulatory factor for maintaining adult β-cell function, whereas MafB plays an essential role in β-cell development during embryogenesis, and its expression in β cells gradually decreases and is restricted to α cells after birth in rodents. However, it was previously observed that MafB started to be reexpressed in insulin-positive (insulin+) β cells in MafA-deficient adult mice. To elucidate how MafB functions in the adult β cell under MafA-deficient conditions, we generated MafA and MafB double-knockout (A0B0) mice in which MafB was specifically deleted from β cells. As a result, the A0B0 mice became more vulnerable to diabetes under a high-fat diet (HFD) treatment, with impaired islet formation and a decreased number of insulin+ β cells because of increased β-cell apoptosis, indicating MafB can take part in the maintenance of adult β cells under certain pathological conditions.
Collapse
|
20
|
Late Cognitive Consequences of Gestational Diabetes to the Offspring, in a New Mouse Model. Mol Neurobiol 2019; 56:7754-7764. [PMID: 31115777 DOI: 10.1007/s12035-019-1624-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/24/2019] [Indexed: 12/17/2022]
Abstract
Gestational diabetes mellitus (GD) is a form of insulin resistance triggered during gestation, which affects approximately 10% of pregnant women. Although previously considered a transient condition with few long-term consequences, growing evidence suggest that GD may be linked to permanent metabolic and neurologic changes in the offspring. Currently available GD models fail to recapitulate the full spectrum of this disease, thus providing limited information about the true burden of this condition. Here, we describe a new mouse model of GD, based on the administration of an insulin receptor antagonist (S961, 30 nmol/kg s.c. daily) during pregnancy. Pregnant mice developed increased fasting glycemia and glucose intolerance in the absence of maternal obesity, with a return to normoglycemia shortly after parturition. Moreover, we showed that the adult offspring of GD dams presented pronounced metabolic and cognitive dysfunction when exposed to short-term high-fat diet (HFD). Our data demonstrate that S961 administration to pregnant mice comprises a valuable approach to study the complex pathophysiology of GD, as well as strategies focused on prevention and treatment of both the mother and the offspring. Our findings suggest that the offspring of GD mothers are more susceptible to metabolic and cognitive impairments when exposed to high-fat diet later in life, thus indicating that approaches to prevent and treat these late effects should be pursued.
Collapse
|
21
|
Banerjee RR. Piecing together the puzzle of pancreatic islet adaptation in pregnancy. Ann N Y Acad Sci 2019; 1411:120-139. [PMID: 29377199 DOI: 10.1111/nyas.13552] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/18/2017] [Accepted: 10/24/2017] [Indexed: 12/20/2022]
Abstract
Pregnancy places acute demands on maternal physiology, including profound changes in glucose homeostasis. Gestation is characterized by an increase in insulin resistance, counterbalanced by an adaptive increase in pancreatic β cell production of insulin. Failure of normal adaptive responses of the islet to increased maternal and fetal demands manifests as gestational diabetes mellitus (GDM). The gestational changes and rapid reversal of islet adaptations following parturition are at least partly driven by an anticipatory program rather than post-factum compensatory adaptations. Here, I provide a comprehensive review of the cellular and molecular mechanisms underlying normal islet adaptation during pregnancy and how dysregulation may lead to GDM. Emerging areas of interest and understudied areas worthy of closer examination in the future are highlighted.
Collapse
Affiliation(s)
- Ronadip R Banerjee
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, and the Comprehensive Diabetes Center, University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| |
Collapse
|
22
|
Dirice E, De Jesus DF, Kahraman S, Basile G, Ng RW, El Ouaamari A, Teo AKK, Bhatt S, Hu J, Kulkarni RN. Human duct cells contribute to β cell compensation in insulin resistance. JCI Insight 2019; 4:99576. [PMID: 30996131 PMCID: PMC6538348 DOI: 10.1172/jci.insight.99576] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/05/2019] [Indexed: 12/19/2022] Open
Abstract
The identification of new sources of β cells is an important endeavor with therapeutic implications for diabetes. Insulin resistance, in physiological states such as pregnancy or in pathological states such as type 2 diabetes (T2D), is characterized by a compensatory increase in β cell mass. To explore the existence of a dynamic β cell reserve, we superimposed pregnancy on the liver-specific insulin receptor-KO (LIRKO) model of insulin resistance that already exhibits β cell hyperplasia and used lineage tracing to track the source of new β cells. Although both control and LIRKO mice displayed increased β cell mass in response to the relative insulin resistance of pregnancy, the further increase in mass in the latter supported a dynamic source that could be traced to pancreatic ducts. Two observations support the translational significance of these findings. First, NOD/SCID-γ LIRKO mice that became pregnant following cotransplantation of human islets and human ducts under the kidney capsule showed enhanced β cell proliferation and an increase in ductal cells positive for transcription factors expressed during β cell development. Second, we identified duct cells positive for immature β cell markers in pancreas sections from pregnant humans and in individuals with T2D. Taken together, during increased insulin demand, ductal cells contribute to the compensatory β cell pool by differentiation/neogenesis.
Collapse
Affiliation(s)
- Ercument Dirice
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Dario F. De Jesus
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
- Graduate Program in Areas of Basic and Applied Biology, Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal
| | - Sevim Kahraman
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Giorgio Basile
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Raymond W.S. Ng
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Abdelfattah El Ouaamari
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Adrian Kee Keong Teo
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Shweta Bhatt
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Jiang Hu
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Rohit N. Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Abdulwahab RA, Alaiya A, Shinwari Z, Allaith AAA, Giha HA. LC‑MS/MS proteomic analysis revealed novel associations of 37 proteins with T2DM and notable upregulation of immunoglobulins. Int J Mol Med 2019; 43:2118-2132. [PMID: 30864687 PMCID: PMC6443330 DOI: 10.3892/ijmm.2019.4127] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/04/2019] [Indexed: 12/29/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a disease associated with a number of metabolic disturbances, including protein metabolism. In the present study, blood samples were obtained from Bahraini subjects, including 6 patients with T2DM and 6 age‑ and sex‑matched, non‑diabetic, healthy controls. Depleted and non‑depleted sera were prepared from the collected blood, and the global protein expression changes were evaluated by liquid chromatography tandem mass spectrometry. Only significantly and markedly differentially‑expressed proteins (P<0.05, analysis of variance; maximum fold change ≥1.5) were considered as candidate proteins for informatics analysis. Accordingly, a total of 62 proteins were identified to be differentially expressed in T2DM, compared with control subjects, and they were grouped functionally into 16 classes of proteins. The largest class was that of the immune‑associated proteins. Additionally, ~25 of these proteins (40%) had previously been associated with DM; however, the association of the other 37 proteins with T2DM was a novel observation. The majority of the identified proteins were upregulated in T2DM. The identified proteins could be involved in the pathogenesis of the disease or serve as disease biomarkers. Further validation of the identified proteins in a large study cohort is required, in order to fully access their potential clinical usefulness.
Collapse
Affiliation(s)
- Rabab Asghar Abdulwahab
- Integrated Science Division, College of Health Sciences, University of Bahrain, Manama 32038, Kingdom of Bahrain
| | - Ayodele Alaiya
- Proteomics Unit, Stem Cell and Tissue Re‑Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Zakia Shinwari
- Proteomics Unit, Stem Cell and Tissue Re‑Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Abdul Ameer A Allaith
- Department of Biology College of Science, University of Bahrain, Zallaq 32038, Kingdom of Bahrain
| | - Hayder A Giha
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama 26671, Kingdom of Bahrain
| |
Collapse
|
24
|
Nteeba J, Kubota K, Wang W, Zhu H, Vivian JL, Dai G, Soares MJ. Pancreatic prolactin receptor signaling regulates maternal glucose homeostasis. J Endocrinol 2019; 241:JOE-18-0518.R2. [PMID: 30798322 PMCID: PMC7189340 DOI: 10.1530/joe-18-0518] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/22/2019] [Indexed: 12/17/2022]
Abstract
Prolactin (PRL) signaling has been implicated in the regulation of glucose homeostatic adaptations to pregnancy. In this report, the PRL receptor (Prlr) gene was conditionally disrupted in the pancreas, creating an animal model which proved useful for investigating the biology and pathology of gestational diabetes including its impacts on fetal and placental development. In mice, pancreatic PRLR signaling was demonstrated to be required for pregnancy-associated changes in maternal β cell mass and function. Disruption of the Prlr gene in the pancreas resulted in fewer insulin producing cells, which failed to expand appropriately during pregnancy resulting in reduced blood insulin levels and maternal glucose intolerance. This inability to sustain normal blood glucose balance during pregnancy worsened with age and a successive pregnancy. The etiology of the insulin insufficiency was attributed to deficits in regulatory pathways controlling β cell development. Additionally, the disturbance in maternal blood glucose homeostasis, was associated with fetal overgrowth and dysregulation of inflammation and prolactin-associated transcripts in the placenta. Overall, these results indicate that the PRLR, acting within the pancreas, mediates maternal pancreatic adaptations to pregnancy and therefore its dysfunction may increase a woman's chances of becoming glucose intolerant during pregnancy.
Collapse
Affiliation(s)
- Jackson Nteeba
- Department of Pathology and Laboratory Medicine, Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kaiyu Kubota
- Department of Pathology and Laboratory Medicine, Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Wenfang Wang
- Department of Clinical Laboratory Sciences, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hao Zhu
- Department of Clinical Laboratory Sciences, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jay L Vivian
- Department of Pathology and Laboratory Medicine, Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Guoli Dai
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Michael J Soares
- Department of Pathology and Laboratory Medicine, Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, Kansas, USA
- Center for Perinatal Research, Children’s Research Institute, Children’s Mercy, Kansas City, Missouri, USA
| |
Collapse
|
25
|
Baeyens L, Lemper M, Staels W, De Groef S, De Leu N, Heremans Y, German MS, Heimberg H. (Re)generating Human Beta Cells: Status, Pitfalls, and Perspectives. Physiol Rev 2018; 98:1143-1167. [PMID: 29717931 DOI: 10.1152/physrev.00034.2016] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus results from disturbed glucose homeostasis due to an absolute (type 1) or relative (type 2) deficiency of insulin, a peptide hormone almost exclusively produced by the beta cells of the endocrine pancreas in a tightly regulated manner. Current therapy only delays disease progression through insulin injection and/or oral medications that increase insulin secretion or sensitivity, decrease hepatic glucose production, or promote glucosuria. These drugs have turned diabetes into a chronic disease as they do not solve the underlying beta cell defects or entirely prevent the long-term complications of hyperglycemia. Beta cell replacement through islet transplantation is a more physiological therapeutic alternative but is severely hampered by donor shortage and immune rejection. A curative strategy should combine newer approaches to immunomodulation with beta cell replacement. Success of this approach depends on the development of practical methods for generating beta cells, either in vitro or in situ through beta cell replication or beta cell differentiation. This review provides an overview of human beta cell generation.
Collapse
Affiliation(s)
- Luc Baeyens
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Marie Lemper
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Willem Staels
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Sofie De Groef
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Nico De Leu
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Yves Heremans
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Michael S German
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| | - Harry Heimberg
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels , Belgium ; Diabetes Center, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, and Department of Medicine, University of California San Francisco , San Francisco, California ; Genentech Safety Assessment, South San Francisco, California ; Investigative Toxicology, UCB BioPharma, Braine-l'Alleud, Belgium ; Department of Pediatrics, Division of Pediatric Endocrinology, Ghent University, Hospital and Department of Pediatrics and Genetics , Ghent , Belgium ; Department of Endocrinology, Universitair Ziekenhuis Brussel, Brussels , Belgium ; and Department of Endocrinology, Algemeen Stedelijk Ziekenhuis Aalst, Aalst, Belgium
| |
Collapse
|
26
|
Moyce BL, Dolinsky VW. Maternal β-Cell Adaptations in Pregnancy and Placental Signalling: Implications for Gestational Diabetes. Int J Mol Sci 2018; 19:ijms19113467. [PMID: 30400566 PMCID: PMC6274918 DOI: 10.3390/ijms19113467] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 10/24/2018] [Accepted: 10/31/2018] [Indexed: 12/14/2022] Open
Abstract
Rates of gestational diabetes mellitus (GDM) are on the rise worldwide, and the number of pregnancies impacted by GDM and resulting complications are also increasing. Pregnancy is a period of unique metabolic plasticity, during which mild insulin resistance is a physiological adaptation to prioritize fetal growth. To compensate for this, the pancreatic β-cell utilizes a variety of adaptive mechanisms, including increasing mass, number and insulin-secretory capacity to maintain glucose homeostasis. When insufficient insulin production does not overcome insulin resistance, hyperglycemia can occur. Changes in the maternal system that occur in GDM such as lipotoxicity, inflammation and oxidative stress, as well as impairments in adipokine and placental signalling, are associated with impaired β-cell adaptation. Understanding these pathways, as well as mechanisms of β-cell dysfunction in pregnancy, can identify novel therapeutic targets beyond diet and lifestyle interventions, insulin and antihyperglycemic agents currently used for treating GDM.
Collapse
Affiliation(s)
- Brittany L Moyce
- Department of Pharmacology & Therapeutics and the Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme of the Children's Hospital Research Institute of Manitoba and the Manitoba Developmental Origins of Chronic Diseases in Children Network (DEVOTION), University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
| | - Vernon W Dolinsky
- Department of Pharmacology & Therapeutics and the Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme of the Children's Hospital Research Institute of Manitoba and the Manitoba Developmental Origins of Chronic Diseases in Children Network (DEVOTION), University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
| |
Collapse
|
27
|
Smith KE, Purvis WG, Davis MA, Min CG, Cooksey AM, Weber CS, Jandova J, Price ND, Molano DS, Stanton JB, Kelly AC, Steyn LV, Lynch RM, Limesand SW, Alexander M, Lakey JRT, Seeberger K, Korbutt GS, Mueller KR, Hering BJ, McCarthy FM, Papas KK. In vitro characterization of neonatal, juvenile, and adult porcine islet oxygen demand, β-cell function, and transcriptomes. Xenotransplantation 2018; 25:e12432. [PMID: 30052287 DOI: 10.1111/xen.12432] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/20/2018] [Accepted: 05/24/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND There is currently a shortage of human donor pancreata which limits the broad application of islet transplantation as a treatment for type 1 diabetes. Porcine islets have demonstrated potential as an alternative source, but a study evaluating islets from different donor ages under unified protocols has yet to be conducted. METHODS Neonatal porcine islets (NPI; 1-3 days), juvenile porcine islets (JPI; 18-21 days), and adult porcine islets (API; 2+ years) were compared in vitro, including assessments of oxygen consumption rate, membrane integrity determined by FDA/PI staining, β-cell proliferation, dynamic glucose-stimulated insulin secretion, and RNA sequencing. RESULTS Oxygen consumption rate normalized to DNA was not significantly different between ages. Membrane integrity was age dependent, and API had the highest percentage of intact cells. API also had the highest glucose-stimulated insulin secretion response during a dynamic insulin secretion assay and had 50-fold higher total insulin content compared to NPI and JPI. NPI and JPI had similar glucose responsiveness, β-cell percentage, and β-cell proliferation rate. Transcriptome analysis was consistent with physiological assessments. API transcriptomes were enriched for cellular metabolic and insulin secretory pathways, while NPI exhibited higher expression of genes associated with proliferation. CONCLUSIONS The oxygen demand, membrane integrity, β-cell function and proliferation, and transcriptomes of islets from API, JPI, and NPI provide a comprehensive physiological comparison for future studies. These assessments will inform the optimal application of each age of porcine islet to expand the availability of islet transplantation.
Collapse
Affiliation(s)
- Kate E Smith
- Department of Physiological Sciences, University of Arizona, Tucson, AZ, USA.,Department of Surgery, University of Arizona, Tucson, AZ, USA
| | | | - Melissa A Davis
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Catherine G Min
- Department of Physiological Sciences, University of Arizona, Tucson, AZ, USA.,Department of Surgery, University of Arizona, Tucson, AZ, USA
| | - Amanda M Cooksey
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Craig S Weber
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Jana Jandova
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | | | - Diana S Molano
- Department of Surgery, University of Arizona, Tucson, AZ, USA
| | | | - Amy C Kelly
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Leah V Steyn
- Department of Surgery, University of Arizona, Tucson, AZ, USA
| | - Ronald M Lynch
- Department of Physiology, University of Arizona, Tucson, AZ, USA
| | - Sean W Limesand
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Michael Alexander
- Department of Surgery, University of California-Irvine, Orange, CA, USA
| | | | - Karen Seeberger
- Department of Surgery, Alberta Diabetes Institute, University of Alberta, Edmonton, AL, Canada
| | - Gregory S Korbutt
- Department of Surgery, Alberta Diabetes Institute, University of Alberta, Edmonton, AL, Canada
| | - Kate R Mueller
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Bernhard J Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Fiona M McCarthy
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | | |
Collapse
|
28
|
Ou K, Zhang J, Jiao Y, Wang ZV, Scherer P, Kaestner KH. Overexpression of ST5, an activator of Ras, has no effect on β-cell proliferation in adult mice. Mol Metab 2018; 11:212-217. [PMID: 29650351 PMCID: PMC6001393 DOI: 10.1016/j.molmet.2018.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 03/15/2018] [Accepted: 03/19/2018] [Indexed: 01/27/2023] Open
Abstract
OBJECTIVE Both Type I and Type II diabetes mellitus result from insufficient functional β-cell mass. Efforts to increase β-cell proliferation as a means to restore β-cell mass have been met with limited success. Suppression of Tumorigenicity 5 (ST5) activates Ras/Erk signaling in the presence of Epidermal Growth Factor (EGF). In the pancreatic islet, Ras/Erk signaling is required for augmented β-cell proliferation during pregnancy, suggesting that ST5 is an appealing candidate to enhance adult β-cell proliferation. We aimed to test the hypothesis that overexpression of ST5 drives adult β-cell proliferation. METHODS We utilized a doxycycline-inducible bitransgenic mouse model to activate β-cell-specific expression of human ST5 in adult mice at will. Islet morphology, β-cell proliferation, and β-cell mass in control and ST5-overexpressing (ST5 OE) animals were analyzed by immunofluorescent staining, under basal and two stimulated metabolic states: pregnancy and streptozotocin (STZ)-induced β-cell loss. RESULTS Doxycycline treatment resulted in robust ST5 overexpression in islets from 12-16 week-old ST5 OE animals compared to controls, without affecting the islet morphology and identity of the β-cells. Under both basal and metabolically stimulated pregnancy states, β-cell proliferation and mass were comparable in ST5 OE and control animals. Furthermore, there was no detectable difference in β-cell proliferation between ST5 OE and control animals in response to STZ-induced β-cell loss. CONCLUSIONS We successfully derived an inducible bitransgenic mouse model to overexpress ST5 specifically in β-cells. However, our findings demonstrate that ST5 overexpression by itself has no mitogenic effect on the adult β-cell under basal and metabolically challenged states.
Collapse
Affiliation(s)
- Kristy Ou
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Jia Zhang
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Yang Jiao
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Zhao V Wang
- Touchstone Diabetes Center, University of Texas Southwestern, Dallas, TX, 75390, USA
| | - Phillipp Scherer
- Touchstone Diabetes Center, University of Texas Southwestern, Dallas, TX, 75390, USA.
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
29
|
Sousa RALDE, Torres YS, Figueiredo CP, Passos GF, Clarke JR. Consequences of gestational diabetes to the brain and behavior of the offspring. AN ACAD BRAS CIENC 2017; 90:2279-2291. [PMID: 28813108 DOI: 10.1590/0001-3765201720170264] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 05/30/2017] [Indexed: 01/11/2023] Open
Abstract
Gestational diabetes mellitus (GD) is a form of insulin resistance triggered during the second/third trimesters of pregnancy in previously normoglycemic women. It is currently estimated that 10% of all pregnancies in the United States show this condition. For many years, the transient nature of GD has led researchers and physicians to assume that long-term consequences were absent. However, GD diagnosis leads to a six-fold increase in the risk of developing type 2 diabetes (T2D) in women and incidence of obesity and T2D is also higher among their infants. Recent and concerning evidences point to detrimental effects of GD on the behavior and cognition of the offspring, which often persist until adolescence or adulthood. Considering that the perinatal period is critical for determination of adult behavior, it is expected that the intra-uterine exposure to hyperglycemia, hyperinsulinemia and pro-inflammatory mediators, hallmark features of GD, might affect brain development. Here, we review early clinical and experimental evidence linking GD to consequences on the behavior of the offspring, focusing on memory and mood disorders. We also discuss initial evidence suggesting that downregulation of insulin signaling cascades are seen in the brains of GD offspring and could contribute to the consequences on their behavior.
Collapse
Affiliation(s)
- Ricardo A L DE Sousa
- School of Pharmacy, Carlos Chagas Filho Street, 373, Building A, Underground, Room 024, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Yasmin S Torres
- School of Pharmacy, Carlos Chagas Filho Street, 373, Building A, Underground, Room 024, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Claudia P Figueiredo
- School of Pharmacy, Carlos Chagas Filho Street, 373, Building A, Underground, Room 024, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Giselle F Passos
- School of Pharmacy, Carlos Chagas Filho Street, 373, Building A, Underground, Room 024, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Julia R Clarke
- School of Pharmacy, Carlos Chagas Filho Street, 373, Building A, Underground, Room 024, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| |
Collapse
|
30
|
Brelje TC, Bhagroo NV, Stout LE, Sorenson RL. Prolactin and oleic acid synergistically stimulate β-cell proliferation and growth in rat islets. Islets 2017; 9:e1330234. [PMID: 28686504 PMCID: PMC5510617 DOI: 10.1080/19382014.2017.1330234] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Islet adaptation to pregnancy is largely influenced by prolactin and placental lactogens. In addition serum lipids are significantly increased. Here, we report the novel observation that prolactin and oleic acid synergistically stimulate islet cell proliferation and islet growth. In neonatal rat islets, prolactin increased proliferation 6-fold, oleic acid 3.5-fold, and their combination 15-fold. The expression of insulin in these dividing cells establishes them as β-cells. Similar changes were seen in islet growth. This synergy is restricted to monounsaturated fatty acids and does not occur with other islet growth factors. Oleic acid increases prolactin-induced STAT5 phosphorylation, even though by itself it is unable to induce STAT5 phosphorylation. Their effects on Erk1/2 phosphorylation are additive. Some of the synergy requires the formation of oleoyl CoA and/or its metabolites. Unexpectedly, methyl oleic acid, a non-metabolizable analog of oleic acid, also shows synergy with prolactin. In summary, prolactin and oleic acid synergistically stimulate islet cell proliferation and islet growth in rat islets, oleic acid increases prolactin-induced STAT5 activation, and requires both the metabolism of oleic acid and non-metabolized oleic acid. Since oleic acid is the most abundant monounsaturated fatty acid in serum that is elevated during pregnancy, it may contribute to increased β-cell proliferation seen during pregnancy.
Collapse
Affiliation(s)
- Todd Clark Brelje
- Department of Genetics, Cell Biology, and Development, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Nicholas V. Bhagroo
- Department of Genetics, Cell Biology, and Development, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Laurence E. Stout
- Department of Genetics, Cell Biology, and Development, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Robert L. Sorenson
- Department of Genetics, Cell Biology, and Development, University of Minnesota Medical School, Minneapolis, MN, USA
- CONTACT Robert L. Sorenson Department of Genetics, Cell Biology, and Development, University of Minnesota Medical School, 6–160 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
31
|
Sordi V, Pellegrini S, Krampera M, Marchetti P, Pessina A, Ciardelli G, Fadini G, Pintus C, Pantè G, Piemonti L. Stem cells to restore insulin production and cure diabetes. Nutr Metab Cardiovasc Dis 2017; 27:583-600. [PMID: 28545927 DOI: 10.1016/j.numecd.2017.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/24/2017] [Accepted: 02/11/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND The advancement of knowledge in the field of regenerative medicine is increasing the therapeutic expectations of patients and clinicians on cell therapy approaches. Within these, stem cell therapies are often evoked as a possible therapeutic option for diabetes, already ongoing or possible in the near future. AIM The purpose of this document is to make a point of the situation on existing knowledge and therapies with stem cells to treat patients with diabetes by focusing on some of the aspects that most frequently raise curiosity and discussion in clinical practice and in the interaction with the patient. In fact, at present there are no clinically approved treatments based on the use of stem cells for the treatment of diabetes, but several therapeutic approaches have already been evaluated or are being evaluated in clinical trials. DATA SYNTHESIS It is possible to identify three large potential application fields: 1) the reconstruction of the β cell mass; 2) the immunomodulation in type 1 diabetes (T1D); 3) the treatment of complications. In this study we will limit the discussion to approaches that have the potential for clinical translation, deliberately omitting aspects of basic biology and preclinical data. Also, we intentionally omit the treatment of the complications that will be the subject of a future document. Finally, an overview of the Italian situation regarding the storage of cord blood cells for the therapy of diabetes will be given.
Collapse
Affiliation(s)
- V Sordi
- Diabetes Research Institute (DRI) - IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - S Pellegrini
- Diabetes Research Institute (DRI) - IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Italy
| | - P Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - A Pessina
- CRC-StaMeTec (Mesenchymal Stem Cells for Cell Therapy), Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - G Ciardelli
- DIMEAS - Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - G Fadini
- Medicine Department (DIMED), University of Padua, Italy
| | - C Pintus
- Italian National Transplant Center (CNT), Italy
| | - G Pantè
- Italian Medicines Agency (AIFA), Italy
| | - L Piemonti
- Diabetes Research Institute (DRI) - IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
32
|
Gao YH, Wu ZX, Xie LQ, Li CX, Mao YQ, Duan YT, Han B, Han SF, Yu Y, Lu HJ, Yang PY, Xu TR, Xia JL, Chen GQ, Wang LS. VHL deficiency augments anthracycline sensitivity of clear cell renal cell carcinomas by down-regulating ALDH2. Nat Commun 2017. [PMID: 28643803 PMCID: PMC5481740 DOI: 10.1038/ncomms15337] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The von Hippel-Lindau (VHL) is deficient in ∼70% of clear-cell renal cell carcinomas (ccRCC), which contributes to the carcinogenesis and drug resistance of ccRCC. Here we show that VHL-deficient ccRCC cells present enhanced cytotoxicity of anthracyclines in a hypoxia-inducible factor-independent manner. By subtractive proteomic analysis coupling with RNAi or overexpression verification, aldehyde dehydrogenase 2 (ALDH2) is found to be transcriptionally regulated by VHL and contributes to enhanced anthracyclines cytotoxicity in ccRCC cells. Furthermore, VHL regulates ALDH2 expression by directly binding the promoter of −130 bp to −160 bp to activate the transcription of hepatocyte nuclear factor 4 alpha (HNF-4α). In addition, a positive correlation is found among the protein expressions of VHL, HNF-4α and ALDH2 in ccRCC samples. These findings will deepen our understanding of VHL function and shed light on precise treatment for ccRCC patients. The VHL tumour suppressor gene is lost in approximately 70% of clear cell renal cell carcinoma (ccRCC). In this study, the authors demonstrate that VHL loss in these tumours augments anthracyclines chemotherapy by down-regulation of ALDH2.
Collapse
Affiliation(s)
- Yao-Hui Gao
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai 201199, China
| | - Zhao-Xia Wu
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai 201199, China
| | - Li-Qi Xie
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai 201199, China
| | - Cai-Xia Li
- Shanghai Universities E-Institute for Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Yu-Qin Mao
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai 201199, China
| | - Yan-Tao Duan
- Shanghai Universities E-Institute for Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Bing Han
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai 201199, China
| | - San-Feng Han
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai 201199, China
| | - Yun Yu
- Shanghai Universities E-Institute for Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Hao-Jie Lu
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai 201199, China
| | - Peng-Yuan Yang
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai 201199, China
| | - Tian-Rui Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Jing-Lin Xia
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai 201199, China
| | - Guo-Qiang Chen
- Shanghai Universities E-Institute for Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| | - Li-Shun Wang
- Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, 170 Xinsong Road, Shanghai 201199, China.,Shanghai Universities E-Institute for Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
| |
Collapse
|
33
|
de Luca A, Hankard R, Borys JM, Sinnett D, Marcil V, Levy E. Nutriepigenomics and malnutrition. Epigenomics 2017; 9:893-917. [DOI: 10.2217/epi-2016-0168] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Epigenetics is defined as the modulation of gene expression without changes to the underlying DNA sequence. Epigenetic alterations, as a consequence of in utero malnutrition, may play a role in susceptibility to develop adulthood diseases and inheritance. However, the mechanistic link between epigenetic modifications and abnormalities in nutrition remains elusive. This review provides an update on the association of suboptimal nutritional environment and the high propensity to produce adult-onset chronic illnesses with a particular focus on modifications in genome functions that occur without alterations to the DNA sequence. We will mention the drivers of the phenotype and pattern of epigenetic markers set down during the reprogramming along with novel preventative and therapeutic strategies. New knowledge of epigenetic alterations is opening a gate toward personalized medicine.
Collapse
Affiliation(s)
- Arnaud de Luca
- Research Center, Sainte-Justine University Hospital Center, Montreal, Quebec H3T 1C5, Canada
- INSERM, U 1069, F-37044 Tours, France
| | - Regis Hankard
- INSERM, U 1069, F-37044 Tours, France
- François Rabelais University, F-37000 Tours, France
| | | | - Daniel Sinnett
- Research Center, Sainte-Justine University Hospital Center, Montreal, Quebec H3T 1C5, Canada
- Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - Valérie Marcil
- Research Center, Sainte-Justine University Hospital Center, Montreal, Quebec H3T 1C5, Canada
- Department of Nutrition, Faculty of Medicine, University of Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Emile Levy
- Research Center, Sainte-Justine University Hospital Center, Montreal, Quebec H3T 1C5, Canada
- EPODE International Network, F-75017 Paris, France
- Department of Nutrition, Faculty of Medicine, University of Montréal, Montreal, Quebec H3T 1J4, Canada
| |
Collapse
|
34
|
Baeyens L, Hindi S, Sorenson RL, German MS. β-Cell adaptation in pregnancy. Diabetes Obes Metab 2016; 18 Suppl 1:63-70. [PMID: 27615133 PMCID: PMC5384851 DOI: 10.1111/dom.12716] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/09/2016] [Indexed: 12/15/2022]
Abstract
Pregnancy in placental mammals places unique demands on the insulin-producing β-cells in the pancreatic islets of Langerhans. The pancreas anticipates the increase in insulin resistance that occurs late in pregnancy by increasing β-cell numbers and function earlier in pregnancy. In rodents, this β-cell expansion depends on secreted placental lactogens that signal through the prolactin receptor. Then at the end of pregnancy, the β-cell population contracts back to its pre-pregnancy size. In the current review, we focus on how glucose metabolism changes during pregnancy, how β-cells anticipate these changes through their response to lactogens and what molecular mechanisms guide the adaptive compensation. In addition, we summarize current knowledge of β-cell adaptation during human pregnancy and what happens when adaptation fails and gestational diabetes ensues. A better understanding of human β-cell adaptation to pregnancy would benefit efforts to predict, prevent and treat gestational diabetes.
Collapse
Affiliation(s)
- L Baeyens
- Diabetes Center, University of California San Francisco, San Francisco
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California San Francisco, San Francisco
| | - S Hindi
- Diabetes Center, University of California San Francisco, San Francisco
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California San Francisco, San Francisco
- Department of Medicine, University of California San Francisco, San Francisco
| | - R L Sorenson
- Department of Genetics, Cell Biology and Development, University of Minnesota Medical School, Minneapolis
| | - M S German
- Diabetes Center, University of California San Francisco, San Francisco.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California San Francisco, San Francisco.
- Department of Medicine, University of California San Francisco, San Francisco.
| |
Collapse
|
35
|
Chellappa K, Deol P, Evans JR, Vuong LM, Chen G, Briançon N, Bolotin E, Lytle C, Nair MG, Sladek FM. Opposing roles of nuclear receptor HNF4α isoforms in colitis and colitis-associated colon cancer. eLife 2016; 5. [PMID: 27166517 PMCID: PMC4907689 DOI: 10.7554/elife.10903] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 05/09/2016] [Indexed: 01/13/2023] Open
Abstract
HNF4α has been implicated in colitis and colon cancer in humans but the role of the different HNF4α isoforms expressed from the two different promoters (P1 and P2) active in the colon is not clear. Here, we show that P1-HNF4α is expressed primarily in the differentiated compartment of the mouse colonic crypt and P2-HNF4α in the proliferative compartment. Exon swap mice that express only P1- or only P2-HNF4α have different colonic gene expression profiles, interacting proteins, cellular migration, ion transport and epithelial barrier function. The mice also exhibit altered susceptibilities to experimental colitis (DSS) and colitis-associated colon cancer (AOM+DSS). When P2-HNF4α-only mice (which have elevated levels of the cytokine resistin-like β, RELMβ, and are extremely sensitive to DSS) are crossed with Retnlb(-/-) mice, they are rescued from mortality. Furthermore, P2-HNF4α binds and preferentially activates the RELMβ promoter. In summary, HNF4α isoforms perform non-redundant functions in the colon under conditions of stress, underscoring the importance of tracking them both in colitis and colon cancer.
Collapse
Affiliation(s)
- Karthikeyani Chellappa
- Department of Cell Biology and Neuroscience, University of California, Riverside, Riverside, United States
| | - Poonamjot Deol
- Department of Cell Biology and Neuroscience, University of California, Riverside, Riverside, United States
| | - Jane R Evans
- Department of Cell Biology and Neuroscience, University of California, Riverside, Riverside, United States
| | - Linh M Vuong
- Department of Cell Biology and Neuroscience, University of California, Riverside, Riverside, United States
| | - Gang Chen
- Division of Biomedical Sciences, University of California, Riverside, Riverside, United States
| | - Nadege Briançon
- Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Eugene Bolotin
- Department of Cell Biology and Neuroscience, University of California, Riverside, Riverside, United States
| | - Christian Lytle
- Division of Biomedical Sciences, University of California, Riverside, Riverside, United States
| | - Meera G Nair
- Division of Biomedical Sciences, University of California, Riverside, Riverside, United States
| | - Frances M Sladek
- Department of Cell Biology and Neuroscience, University of California, Riverside, Riverside, United States
| |
Collapse
|
36
|
Guan B, Li W, Li F, Xie Y, Ni Q, Gu Y, Li X, Wang Q, Zhang H, Ning G. Sfrp5 mediates glucose-induced proliferation in rat pancreatic β-cells. J Endocrinol 2016; 229:73-83. [PMID: 26869333 DOI: 10.1530/joe-15-0535] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 02/11/2016] [Indexed: 01/07/2023]
Abstract
The cellular and molecular mechanisms of glucose-stimulated β-cell proliferation are poorly understood. Recently, secreted frizzled-related protein 5 (encoded by Sfrp5; a Wnt signaling inhibitor) has been demonstrated to be involved in β-cell proliferation in obesity. A previous study demonstrated that glucose enhanced Wnt signaling to promote cell proliferation. We hypothesized that inhibition of SFRP5 contributes to glucose-stimulated β-cell proliferation. In this study, we found that the Sfrp5 level was significantly reduced in high glucose-treated INS-1 cells, primary rat β-cells, and islets isolated from glucose-infused rats. Overexpression of SFRP5 diminished glucose-stimulated proliferation in both INS-1 cells and primary β-cells, with a concomitant inhibition of the Wnt signaling pathway and decreased cyclin D2 expression. In addition, we showed that glucose-induced Sfrp5 suppression was modulated by the PI3K/AKT pathway. Therefore, we conclude that glucose inhibits Sfrp5 expression via the PI3K/AKT pathway and hence promotes rat pancreatic β-cell proliferation.
Collapse
Affiliation(s)
- Binbin Guan
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China Department of EndocrinologyFuJian Union hospital, Fuzhou, P R China
| | - Wenyi Li
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Fengying Li
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Yun Xie
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Qicheng Ni
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Yanyun Gu
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Xiaoying Li
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Qidi Wang
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Hongli Zhang
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| | - Guang Ning
- Shanghai Institute of Endocrine and Metabolic DiseasesShanghai Clinical Center for Endocrine and Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P R China
| |
Collapse
|
37
|
Robitaille K, Rourke JL, McBane JE, Fu A, Baird S, Du Q, Kin T, Shapiro AMJ, Screaton RA. High-throughput Functional Genomics Identifies Regulators of Primary Human Beta Cell Proliferation. J Biol Chem 2016; 291:4614-25. [PMID: 26740620 PMCID: PMC4813485 DOI: 10.1074/jbc.m115.683912] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Indexed: 02/05/2023] Open
Abstract
The expansion of cells for regenerative therapy will require the genetic dissection of complex regulatory mechanisms governing the proliferation of non-transformed human cells. Here, we report the development of a high-throughput RNAi screening strategy specifically for use in primary cells and demonstrate that silencing the cell cycle-dependent kinase inhibitors CDKN2C/p18 or CDKN1A/p21 facilitates cell cycle entry of quiescent adult human pancreatic beta cells. This work identifies p18 and p21 as novel targets for promoting proliferation of human beta cells and demonstrates the promise of functional genetic screens for dissecting therapeutically relevant state changes in primary human cells.
Collapse
Affiliation(s)
- Karine Robitaille
- From the Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1
| | | | - Joanne E McBane
- From the Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1
| | - Accalia Fu
- From the Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, the Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario K1H 8M5
| | - Stephen Baird
- From the Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1
| | - Qiujiang Du
- the Sunnybrook Research Institute, Toronto, Ontario M4N 3M5
| | - Tatsuya Kin
- the Clinical Islet Transplant Program, University of Alberta, 8215-112 Street, Edmonton, Alberta T6G 2C8, and
| | - A M James Shapiro
- the Clinical Islet Transplant Program, University of Alberta, 8215-112 Street, Edmonton, Alberta T6G 2C8, and
| | - Robert A Screaton
- the Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, the Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
38
|
Sheaffer KL, Elliott EN, Kaestner KH. DNA Hypomethylation Contributes to Genomic Instability and Intestinal Cancer Initiation. Cancer Prev Res (Phila) 2016; 9:534-46. [PMID: 26883721 DOI: 10.1158/1940-6207.capr-15-0349] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/08/2016] [Indexed: 12/12/2022]
Abstract
Intestinal cancer is a heterogeneous disease driven by genetic mutations and epigenetic changes. Approximately 80% of sporadic colorectal cancers are initiated by mutation and inactivation of the adenomatous polyposis coli (APC) gene, which results in unrestrained intestinal epithelial growth and formation of adenomas. Aberrant DNA methylation promotes cancer progression by the inactivation of tumor suppressor genes via promoter methylation. In addition, global DNA hypomethylation is often seen before the formation of adenomas, suggesting that it contributes to neoplastic transformation. Previous studies employed mice with a hypomorphic mutation in DNA methyltransferase 1 (Dnmt1), which exhibited constitutive global DNA hypomethylation and decreased tumorigenesis in the Apc(Min/+) mouse model of intestinal cancer. However, the consequences of intestinal epithelial-specific acute hypomethylation during Apc(Min/+) tumor initiation have not been reported. Using temporally controlled intestinal epithelial-specific gene ablation, we show that total loss of Dnmt1 in the Apc(Min/+) mouse model of intestinal cancer causes accelerated adenoma initiation. Deletion of Dnmt1 precipitates an acute response characterized by hypomethylation of repetitive elements and genomic instability, which surprisingly is followed by remethylation with time. Two months post-Dnmt1 ablation, mice display increased macroadenoma load, consistent with a role for Dnmt1 and DNA methylation in maintaining genomic stability. These data suggest that DNA hypomethylation plays a previously unappreciated role in intestinal adenoma initiation. Cancer Prev Res; 9(7); 534-46. ©2016 AACRSee related article by Lee and Laird, p. 509.
Collapse
Affiliation(s)
- Karyn L Sheaffer
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ellen N Elliott
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
39
|
Elliott EN, Sheaffer KL, Kaestner KH. The 'de novo' DNA methyltransferase Dnmt3b compensates the Dnmt1-deficient intestinal epithelium. eLife 2016; 5:e12975. [PMID: 26808831 PMCID: PMC4786433 DOI: 10.7554/elife.12975] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/25/2016] [Indexed: 12/11/2022] Open
Abstract
Dnmt1 is critical for immediate postnatal intestinal development, but is not required for the survival of the adult intestinal epithelium, the only rapidly dividing somatic tissue for which this has been shown. Acute Dnmt1 deletion elicits dramatic hypomethylation and genomic instability. Recovery of DNA methylation state and intestinal health is dependent on the de novo methyltransferase Dnmt3b. Ablation of both Dnmt1 and Dnmt3b in the intestinal epithelium is lethal, while deletion of either Dnmt1 or Dnmt3b has no effect on survival. These results demonstrate that Dnmt1 and Dnmt3b cooperate to maintain DNA methylation and genomic integrity in the intestinal epithelium.
Collapse
Affiliation(s)
- Ellen N Elliott
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Karyn L Sheaffer
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Klaus H Kaestner
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
40
|
Horn S, Kirkegaard JS, Hoelper S, Seymour PA, Rescan C, Nielsen JH, Madsen OD, Jensen JN, Krüger M, Grønborg M, Ahnfelt-Rønne J. Research Resource: A Dual Proteomic Approach Identifies Regulated Islet Proteins During β-Cell Mass Expansion In Vivo. Mol Endocrinol 2015; 30:133-43. [PMID: 26649805 DOI: 10.1210/me.2015-1208] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Diabetes is characterized by insulin insufficiency due to a relative paucity of functional β-cell mass. Thus, strategies for increasing β-cell mass in situ are sought-after for therapeutic purposes. Pregnancy is a physiological state capable of inducing robust β-cell mass expansion, however, the mechanisms driving this expansion are not fully understood. Thus, the aim of this study was to characterize pregnancy-induced changes in the islet proteome at the peak of β-cell proliferation in mice. Islets from pregnant and nonpregnant littermates were compared via 2 proteomic strategies. In vivo pulsed stable isotope labeling of amino acids in cell culture was used to monitor de novo protein synthesis during the first 14.5 days of pregnancy. In parallel, protein abundance was determined using ex vivo dimethyl labelling at gestational day 14.5. Comparison of the 2 datasets revealed 170 islet proteins to be up regulated as a response to pregnancy. These included several proteins, not previously associated with pregnancy-induced islet expansion, such as CLIC1, STMN1, MCM6, PPIB, NEDD4, and HLTF. Confirming the validity of our approach, we also identified proteins encoded by genes known to be associated with pregnancy-induced islet expansion, such as CHGB, IGFBP5, MATN2, EHHADH, IVD, and BMP1. Bioinformatic analyses demonstrated enrichment and activation of the biological functions: "protein synthesis" and "proliferation," and predicted the transcription factors HNF4α, MYC, MYCN, E2F1, NFE2L2, and HNF1α as upstream regulators of the observed expressional changes. As the first characterization of the islet-proteome during pregnancy, this study provides novel insight into the mechanisms involved in promoting pregnancy-induced β-cell mass expansion and function.
Collapse
Affiliation(s)
- Signe Horn
- Global Research (S.Hor., J.S.K., C.R., O.D.M., J.N.J., M.G., J.A.-R.), Novo Nordisk A/S, 2870 Maaloev, Denmark; Department of Biomedical Sciences (S.Hor., J.S.K., J.H.N.), University of Copenhagen, 2200 Copenhagen N, Denmark; Max Planck Institute for Heart and Lung Research (S.Hoe.), 61231 Bad Nauheim, Germany; The Danish Stem Cell Center (P.A.S., O.D.M.), University of Copenhagen, 2200 Copenhagen N, Denmark; and Institute of Genetics (M.K.), Cluster of Excellence in Cellular Stress Responses, University of Cologne, 50931 Cologne, Germany
| | - Jeannette S Kirkegaard
- Global Research (S.Hor., J.S.K., C.R., O.D.M., J.N.J., M.G., J.A.-R.), Novo Nordisk A/S, 2870 Maaloev, Denmark; Department of Biomedical Sciences (S.Hor., J.S.K., J.H.N.), University of Copenhagen, 2200 Copenhagen N, Denmark; Max Planck Institute for Heart and Lung Research (S.Hoe.), 61231 Bad Nauheim, Germany; The Danish Stem Cell Center (P.A.S., O.D.M.), University of Copenhagen, 2200 Copenhagen N, Denmark; and Institute of Genetics (M.K.), Cluster of Excellence in Cellular Stress Responses, University of Cologne, 50931 Cologne, Germany
| | - Soraya Hoelper
- Global Research (S.Hor., J.S.K., C.R., O.D.M., J.N.J., M.G., J.A.-R.), Novo Nordisk A/S, 2870 Maaloev, Denmark; Department of Biomedical Sciences (S.Hor., J.S.K., J.H.N.), University of Copenhagen, 2200 Copenhagen N, Denmark; Max Planck Institute for Heart and Lung Research (S.Hoe.), 61231 Bad Nauheim, Germany; The Danish Stem Cell Center (P.A.S., O.D.M.), University of Copenhagen, 2200 Copenhagen N, Denmark; and Institute of Genetics (M.K.), Cluster of Excellence in Cellular Stress Responses, University of Cologne, 50931 Cologne, Germany
| | - Philip A Seymour
- Global Research (S.Hor., J.S.K., C.R., O.D.M., J.N.J., M.G., J.A.-R.), Novo Nordisk A/S, 2870 Maaloev, Denmark; Department of Biomedical Sciences (S.Hor., J.S.K., J.H.N.), University of Copenhagen, 2200 Copenhagen N, Denmark; Max Planck Institute for Heart and Lung Research (S.Hoe.), 61231 Bad Nauheim, Germany; The Danish Stem Cell Center (P.A.S., O.D.M.), University of Copenhagen, 2200 Copenhagen N, Denmark; and Institute of Genetics (M.K.), Cluster of Excellence in Cellular Stress Responses, University of Cologne, 50931 Cologne, Germany
| | - Claude Rescan
- Global Research (S.Hor., J.S.K., C.R., O.D.M., J.N.J., M.G., J.A.-R.), Novo Nordisk A/S, 2870 Maaloev, Denmark; Department of Biomedical Sciences (S.Hor., J.S.K., J.H.N.), University of Copenhagen, 2200 Copenhagen N, Denmark; Max Planck Institute for Heart and Lung Research (S.Hoe.), 61231 Bad Nauheim, Germany; The Danish Stem Cell Center (P.A.S., O.D.M.), University of Copenhagen, 2200 Copenhagen N, Denmark; and Institute of Genetics (M.K.), Cluster of Excellence in Cellular Stress Responses, University of Cologne, 50931 Cologne, Germany
| | - Jens H Nielsen
- Global Research (S.Hor., J.S.K., C.R., O.D.M., J.N.J., M.G., J.A.-R.), Novo Nordisk A/S, 2870 Maaloev, Denmark; Department of Biomedical Sciences (S.Hor., J.S.K., J.H.N.), University of Copenhagen, 2200 Copenhagen N, Denmark; Max Planck Institute for Heart and Lung Research (S.Hoe.), 61231 Bad Nauheim, Germany; The Danish Stem Cell Center (P.A.S., O.D.M.), University of Copenhagen, 2200 Copenhagen N, Denmark; and Institute of Genetics (M.K.), Cluster of Excellence in Cellular Stress Responses, University of Cologne, 50931 Cologne, Germany
| | - Ole D Madsen
- Global Research (S.Hor., J.S.K., C.R., O.D.M., J.N.J., M.G., J.A.-R.), Novo Nordisk A/S, 2870 Maaloev, Denmark; Department of Biomedical Sciences (S.Hor., J.S.K., J.H.N.), University of Copenhagen, 2200 Copenhagen N, Denmark; Max Planck Institute for Heart and Lung Research (S.Hoe.), 61231 Bad Nauheim, Germany; The Danish Stem Cell Center (P.A.S., O.D.M.), University of Copenhagen, 2200 Copenhagen N, Denmark; and Institute of Genetics (M.K.), Cluster of Excellence in Cellular Stress Responses, University of Cologne, 50931 Cologne, Germany
| | - Jan N Jensen
- Global Research (S.Hor., J.S.K., C.R., O.D.M., J.N.J., M.G., J.A.-R.), Novo Nordisk A/S, 2870 Maaloev, Denmark; Department of Biomedical Sciences (S.Hor., J.S.K., J.H.N.), University of Copenhagen, 2200 Copenhagen N, Denmark; Max Planck Institute for Heart and Lung Research (S.Hoe.), 61231 Bad Nauheim, Germany; The Danish Stem Cell Center (P.A.S., O.D.M.), University of Copenhagen, 2200 Copenhagen N, Denmark; and Institute of Genetics (M.K.), Cluster of Excellence in Cellular Stress Responses, University of Cologne, 50931 Cologne, Germany
| | - Marcus Krüger
- Global Research (S.Hor., J.S.K., C.R., O.D.M., J.N.J., M.G., J.A.-R.), Novo Nordisk A/S, 2870 Maaloev, Denmark; Department of Biomedical Sciences (S.Hor., J.S.K., J.H.N.), University of Copenhagen, 2200 Copenhagen N, Denmark; Max Planck Institute for Heart and Lung Research (S.Hoe.), 61231 Bad Nauheim, Germany; The Danish Stem Cell Center (P.A.S., O.D.M.), University of Copenhagen, 2200 Copenhagen N, Denmark; and Institute of Genetics (M.K.), Cluster of Excellence in Cellular Stress Responses, University of Cologne, 50931 Cologne, Germany
| | - Mads Grønborg
- Global Research (S.Hor., J.S.K., C.R., O.D.M., J.N.J., M.G., J.A.-R.), Novo Nordisk A/S, 2870 Maaloev, Denmark; Department of Biomedical Sciences (S.Hor., J.S.K., J.H.N.), University of Copenhagen, 2200 Copenhagen N, Denmark; Max Planck Institute for Heart and Lung Research (S.Hoe.), 61231 Bad Nauheim, Germany; The Danish Stem Cell Center (P.A.S., O.D.M.), University of Copenhagen, 2200 Copenhagen N, Denmark; and Institute of Genetics (M.K.), Cluster of Excellence in Cellular Stress Responses, University of Cologne, 50931 Cologne, Germany
| | - Jonas Ahnfelt-Rønne
- Global Research (S.Hor., J.S.K., C.R., O.D.M., J.N.J., M.G., J.A.-R.), Novo Nordisk A/S, 2870 Maaloev, Denmark; Department of Biomedical Sciences (S.Hor., J.S.K., J.H.N.), University of Copenhagen, 2200 Copenhagen N, Denmark; Max Planck Institute for Heart and Lung Research (S.Hoe.), 61231 Bad Nauheim, Germany; The Danish Stem Cell Center (P.A.S., O.D.M.), University of Copenhagen, 2200 Copenhagen N, Denmark; and Institute of Genetics (M.K.), Cluster of Excellence in Cellular Stress Responses, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
41
|
Nilsson E, Matte A, Perfilyev A, de Mello VD, Käkelä P, Pihlajamäki J, Ling C. Epigenetic Alterations in Human Liver From Subjects With Type 2 Diabetes in Parallel With Reduced Folate Levels. J Clin Endocrinol Metab 2015; 100:E1491-501. [PMID: 26418287 PMCID: PMC4702449 DOI: 10.1210/jc.2015-3204] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Epigenetic variation may contribute to the development of complex metabolic diseases such as type 2 diabetes (T2D). Hepatic insulin resistance is a hallmark of T2D. However, it remains unknown whether epigenetic alterations take place in the liver from diabetic subjects. Therefore, we investigated the genome-wide DNA methylation pattern in the liver from subjects with T2D and nondiabetic controls and related epigenetic alterations to gene expression and circulating folate levels. RESEARCH DESIGN AND METHODS Liver biopsies were obtained from 35 diabetic and 60 nondiabetic subjects, which are part of the Kuopio Obesity Surgery Study. The genome-wide DNA methylation pattern was analyzed in the liver using the HumanMethylation450 BeadChip. RNA expression was analyzed from a subset of subjects using the HumanHT-12 Expression BeadChip. RESULTS After correction for multiple testing, we identified 251 individual CpG sites that exhibit differential DNA methylation in liver obtained from T2D compared with nondiabetic subjects (Q < .05). These include CpG sites annotated to genes that are biologically relevant to the development of T2D such as GRB10, ABCC3, MOGAT1, and PRDM16. The vast majority of the significant CpG sites (94%) displayed decreased DNA methylation in liver from subjects with T2D. The hypomethylation found in liver from diabetic subjects may be explained by reduced folate levels. Indeed, subjects with T2D had significantly reduced erythrocyte folate levels compared with nondiabetic subjects. We further identified 29 genes that displayed both differential DNA methylation and gene expression in human T2D liver including the imprinted gene H19. CONCLUSIONS Our study highlights the importance of epigenetic and transcriptional changes in the liver from subjects with T2D. Reduced circulating folate levels may provide an explanation for hypomethylation in the human diabetic liver.
Collapse
|
42
|
Thyroid-Stimulating Hormone Increases HNF-4α Phosphorylation via cAMP/PKA Pathway in the Liver. Sci Rep 2015; 5:13409. [PMID: 26302721 PMCID: PMC4548215 DOI: 10.1038/srep13409] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 07/27/2015] [Indexed: 12/16/2022] Open
Abstract
Hepatocyte nuclear factor-4 alpha (HNF-4α) is an orphan nuclear receptor with important roles in hepatic metabolism. Protein phosphorylation plays a functional role in its nuclear localization, DNA binding, and transactivation. Thyroid-stimulating hormone (TSH) is a hormone produced by the anterior pituitary gland, whose direct effect on the metabolic pathway has been observed. Our previous study demonstrated that TSH significantly decreases hepatic nuclear HNF-4α expression. However, whether TSH can influence HNF-4α phosphorylation is unclear. Here, we discovered that TSH can increase HNF-4α phosphorylation and modulate its subcellularlocalization. When HepG2 cells were treated with TSH, the phosphorylation of HNF-4α increased and its nuclear localization was interrupted. Cytoplasmic HNF-4α increased, while nuclear HNF-4α decreased. When the cAMP/PKA pathway was inhibited by the PKA inhibitor H89 and the adenylate cyclase (AC) inhibitor SQ22536, the TSH-mediated phosphorylation of HNF-4α was disrupted. When Tshr was silenced in mice, the phosphorylation of HNF-4α decreased, and cytoplasmic HNF-4α decreased while nuclear HNF-4α increased. In conclusion, our study revealed a novel mechanism by which TSH regulated the hepatic HNF-4α subcellular localization, suggesting the possibility that one of the effects of TSH is to reduce the expression of HNF-4α target genes.
Collapse
|
43
|
Stewart AF, Hussain MA, García-Ocaña A, Vasavada RC, Bhushan A, Bernal-Mizrachi E, Kulkarni RN. Human β-cell proliferation and intracellular signaling: part 3. Diabetes 2015; 64:1872-85. [PMID: 25999530 PMCID: PMC4439562 DOI: 10.2337/db14-1843] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This is the third in a series of Perspectives on intracellular signaling pathways coupled to proliferation in pancreatic β-cells. We contrast the large knowledge base in rodent β-cells with the more limited human database. With the increasing incidence of type 1 diabetes and the recognition that type 2 diabetes is also due in part to a deficiency of functioning β-cells, there is great urgency to identify therapeutic approaches to expand human β-cell numbers. Therapeutic approaches might include stem cell differentiation, transdifferentiation, or expansion of cadaver islets or residual endogenous β-cells. In these Perspectives, we focus on β-cell proliferation. Past Perspectives reviewed fundamental cell cycle regulation and its upstream regulation by insulin/IGF signaling via phosphatidylinositol-3 kinase/mammalian target of rapamycin signaling, glucose, glycogen synthase kinase-3 and liver kinase B1, protein kinase Cζ, calcium-calcineurin-nuclear factor of activated T cells, epidermal growth factor/platelet-derived growth factor family members, Wnt/β-catenin, leptin, and estrogen and progesterone. Here, we emphasize Janus kinase/signal transducers and activators of transcription, Ras/Raf/extracellular signal-related kinase, cadherins and integrins, G-protein-coupled receptors, and transforming growth factor β signaling. We hope these three Perspectives will serve to introduce these pathways to new researchers and will encourage additional investigators to focus on understanding how to harness key intracellular signaling pathways for therapeutic human β-cell regeneration for diabetes.
Collapse
Affiliation(s)
- Andrew F Stewart
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mehboob A Hussain
- Departments of Medicine and Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD
| | - Adolfo García-Ocaña
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rupangi C Vasavada
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Anil Bhushan
- Diabetes Center, University of California, San Francisco, San Francisco, CA
| | - Ernesto Bernal-Mizrachi
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan, Ann Arbor, MI, and VA Ann Arbor Healthcare System, Ann Arbor, MI
| | - Rohit N Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| |
Collapse
|
44
|
Ye R, Wang M, Wang QA, Scherer PE. Adiponectin-mediated antilipotoxic effects in regenerating pancreatic islets. Endocrinology 2015; 156:2019-28. [PMID: 25815422 PMCID: PMC4430619 DOI: 10.1210/en.2015-1066] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pathways that stimulate β-cell regeneration remain of great clinical interest, yet effective therapeutic avenues that promote survival or reconstitution of β-cell mass remain elusive. Using a mouse model with inducible β-cell apoptosis followed by adiponectin-mediated regeneration, we aimed to identify key molecules boosting β-cell viability. In the regenerating pancreatic islets, we examined changes within the transcriptome and observed an extensive up-regulation of genes encoding proteins involved in lipid transport and metabolism. The most prominent targets were further confirmed by quantitative PCR and immunofluorescence. Among the upstream regulators predicted by pathway analysis of the transcriptome, we detected enhanced levels of 2 key transcription factors, Hepatocyte Nuclear Factor 4α and Peroxisome Proliferator-Activated Receptorα. Our data suggest that improving pancreatic islet lipid metabolism as an important antilipotoxic phenomenon to boost β-cell regeneration. This is primarily mediated by the adipokine adiponectin that exerts its action on both the beta-cell directly as well as on the adipocyte. Adiponectin induces lipid metabolism gene expression in regenerating islets through Hepatocyte Nuclear Factor 4α and Peroxisome Proliferator-Activated Receptorα. Adiponectin also modulates leptin levels via preserving adipose tissue mass in the insulinopenic state.
Collapse
Affiliation(s)
- Risheng Ye
- Touchstone Diabetes Center (R.Y., Q.A.W., P.E.S.), Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390; Hamon Center for Therapeutic Oncology Research (M.W.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390; and Department of Cell Biology (P.E.S.), The University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | | | | | | |
Collapse
|
45
|
Elliott EN, Sheaffer KL, Schug J, Stappenbeck TS, Kaestner KH. Dnmt1 is essential to maintain progenitors in the perinatal intestinal epithelium. Development 2015; 142:2163-72. [PMID: 26023099 DOI: 10.1242/dev.117341] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 05/05/2015] [Indexed: 11/20/2022]
Abstract
The DNA methyltransferase Dnmt1 maintains DNA methylation patterns and genomic stability in several in vitro cell systems. Ablation of Dnmt1 in mouse embryos causes death at the post-gastrulation stage; however, the functions of Dnmt1 and DNA methylation in organogenesis remain unclear. Here, we report that Dnmt1 is crucial during perinatal intestinal development. Loss of Dnmt1 in intervillus progenitor cells causes global hypomethylation, DNA damage, premature differentiation, apoptosis and, consequently, loss of nascent villi. We further confirm the crucial role of Dnmt1 during crypt development using the in vitro organoid culture system, and illustrate a clear differential requirement for Dnmt1 in immature versus mature organoids. These results demonstrate an essential role for Dnmt1 in maintaining genomic stability during intestinal development and the establishment of intestinal crypts.
Collapse
Affiliation(s)
- Ellen N Elliott
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karyn L Sheaffer
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan Schug
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thaddeus S Stappenbeck
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
46
|
Ginseng berry extract supplementation improves age-related decline of insulin signaling in mice. Nutrients 2015; 7:3038-53. [PMID: 25912041 PMCID: PMC4425188 DOI: 10.3390/nu7043038] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 03/23/2015] [Accepted: 04/10/2015] [Indexed: 11/20/2022] Open
Abstract
The aim of this study was to evaluate the effects of ginseng berry extract on insulin sensitivity and associated molecular mechanisms in aged mice. C57BL/6 mice (15 months old) were maintained on a regular diet (CON) or a regular diet supplemented with 0.05% ginseng berry extract (GBD) for 24 or 32 weeks. GBD-fed mice showed significantly lower serum insulin levels (p = 0.016) and insulin resistance scores (HOMA-IR) (p = 0.012), suggesting that GBD improved insulin sensitivity. Pancreatic islet hypertrophy was also ameliorated in GBD-fed mice (p = 0.007). Protein levels of tyrosine phosphorylated insulin receptor substrate (IRS)-1 (p = 0.047), and protein kinase B (AKT) (p = 0.037), were up-regulated in the muscle of insulin-injected GBD-fed mice compared with CON-fed mice. The expressions of forkhead box protein O1 (FOXO1) (p = 0.036) and peroxisome proliferator-activated receptor gamma (PPARγ) (p = 0.032), which are known as aging- and insulin resistance-related genes, were also increased in the muscle of GBD-fed mice. We conclude that ginseng berry extract consumption might increase activation of IRS-1 and AKT, contributing to the improvement of insulin sensitivity in aged mice.
Collapse
|
47
|
Yamagata K. Roles of HNF1α and HNF4α in pancreatic β-cells: lessons from a monogenic form of diabetes (MODY). VITAMINS AND HORMONES 2015; 95:407-23. [PMID: 24559927 DOI: 10.1016/b978-0-12-800174-5.00016-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mutations in the genes encoding hepatocyte nuclear factor (HNF)1α and HNF4α cause a monogenic form of diabetes mellitus known as maturity-onset diabetes of the young (MODY). The primary cause of MODY is an impairment of glucose-stimulated insulin secretion by pancreatic β-cells, indicating the important roles of HNF1α and HNF4α in β-cells. Large-scale genetic studies have clarified that the common variants of HNF1α and HNF4α genes are also associated with type 2 diabetes, suggesting that they are involved in the pathogenesis of both diseases. Recent experimental studies revealed that HNF1α controls both β-cell function and growth by regulating target genes such as glucose transporter 2, pyruvate kinase, collectrin, hepatocyte growth factor activator, and HNF4α. In contrast, HNF4α mainly regulates the function of β-cells. Although direct target genes of HNF4α in β-cells are largely unknown, we recently identified Anks4b as a novel target of HNF4α that regulates β-cell susceptibility to endoplasmic reticulum stress. Studies of MODY have led to a better understanding of the molecular mechanism of glucose-stimulated insulin secretion by pancreatic β-cells.
Collapse
Affiliation(s)
- Kazuya Yamagata
- Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
48
|
Sangan CB, Jover R, Heimberg H, Tosh D. In vitro reprogramming of pancreatic alpha cells towards a beta cell phenotype following ectopic HNF4α expression. Mol Cell Endocrinol 2015; 399:50-9. [PMID: 25224487 DOI: 10.1016/j.mce.2014.09.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 08/21/2014] [Accepted: 09/09/2014] [Indexed: 12/31/2022]
Abstract
There is currently a shortage of organ donors available for pancreatic beta cell transplantation into diabetic patients. An alternative source of beta cells is pre-existing pancreatic cells. While we know that beta cells can arise directly from alpha cells during pancreatic regeneration we do not understand the molecular basis for the switch in phenotype. The aim of the present study was to investigate if hepatocyte nuclear factor 4 alpha (HNF4α), a transcription factor essential for a normal beta cell phenotype, could induce the reprogramming of alpha cells towards potential beta cells. We utilised an in vitro model of pancreatic alpha cells, the murine αTC1-9 cell line. We initially characterised the αTC1-9 cell line before and following adenovirus-mediated ectopic expression of HNF4α. We analysed the phenotype at transcript and protein level and assessed its glucose-responsiveness. Ectopic HNF4α expression in the αTC1-9 cell line induced a change in morphology (1.7-fold increase in size), suppressed glucagon expression, induced key beta cell-specific markers (insulin, C-peptide, glucokinase, GLUT2 and Pax4) and pancreatic polypeptide (PP) and enabled the cells to secrete insulin in a glucose-regulated manner. In conclusion, HNF4α reprograms alpha cells to beta-like cells.
Collapse
Affiliation(s)
| | - Ramiro Jover
- Experimental Hepatology Unit. Hosp. La Fe & Dep. Biochemistry, University of Valencia. CIBERehd, Spain
| | - Harry Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - David Tosh
- Centre for Regenerative Medicine, University of Bath, Bath, UK.
| |
Collapse
|
49
|
Alejandro EU, Gregg B, Blandino-Rosano M, Cras-Méneur C, Bernal-Mizrachi E. Natural history of β-cell adaptation and failure in type 2 diabetes. Mol Aspects Med 2014; 42:19-41. [PMID: 25542976 DOI: 10.1016/j.mam.2014.12.002] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 11/04/2014] [Accepted: 12/05/2014] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes mellitus (T2D) is a complex disease characterized by β-cell failure in the setting of insulin resistance. The current evidence suggests that genetic predisposition, and environmental factors can impair the capacity of the β-cells to respond to insulin resistance and ultimately lead to their failure. However, genetic studies have demonstrated that known variants account for less than 10% of the overall estimated T2D risk, suggesting that additional unidentified factors contribute to susceptibility of this disease. In this review, we will discuss the different stages that contribute to the development of β-cell failure in T2D. We divide the natural history of this process in three major stages: susceptibility, β-cell adaptation and β-cell failure, and provide an overview of the molecular mechanisms involved. Further research into mechanisms will reveal key modulators of β-cell failure and thus identify possible novel therapeutic targets and potential interventions to protect against β-cell failure.
Collapse
Affiliation(s)
- Emilyn U Alejandro
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA
| | - Brigid Gregg
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Manuel Blandino-Rosano
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA
| | - Corentin Cras-Méneur
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA
| | - Ernesto Bernal-Mizrachi
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA; VA Ann Arbor Healthcare System, Ann Arbor, MI, USA.
| |
Collapse
|
50
|
Manyes L, Arribas M, Gomez C, Calzada N, Fernandez-Medarde A, Santos E. Transcriptional profiling reveals functional links between RasGrf1 and Pttg1 in pancreatic beta cells. BMC Genomics 2014; 15:1019. [PMID: 25421944 PMCID: PMC4301450 DOI: 10.1186/1471-2164-15-1019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 11/06/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Our prior characterization of RasGrf1 deficient mice uncovered significant defects in pancreatic islet count and size as well as beta cell development and signaling function, raising question about the mechanisms linking RasGrf1 to the generation of those "pancreatic" phenotypes. RESULTS Here, we compared the transcriptional profile of highly purified pancreatic islets from RasGrf1 KO mice to that of WT control animals using commercial oligonucleotide microarrays. RasGrf1 elimination resulted in differential gene expression of numerous components of MAPK- and Calcium-signaling pathways, suggesting a relevant contribution of this GEF to modulation of cellular signaling in the cell lineages integrating the pancreatic islets. Whereas the overall transcriptional profile of pancreatic islets was highly specific in comparison to other organs of the same KO mice, a significant specific repression of Pttg1 was a common transcriptional alteration shared with other tissues of neuroectodermal origin. This observation, together with the remarkable pancreatic phenotypic similarities between RasGrf1 KO and Pttg1 KO mice suggested the possibility of proximal functional regulatory links between RasGrf1 and Pttg1 in pancreatic cell lineages expressing these proteins.Analysis of the mPttg1 promoter region identified specific recognition sites for numerous transcription factors which were also found to be differentially expressed in RasGrf1 KO pancreatic islets and are known to be relevant for Ras-ERK signaling as well as beta cell function. Reporter luciferase assays in BT3 insulinoma cells demonstrated the ability of RasGrf1 to modulate mPttg1 promoter activity through ERK-mediated signals. Analysis of the phenotypic interplay between RasGrf1 and Pttg1 in double knockout RasGrf1/Pttg1 mice showed that combined elimination of the two loci resulted in dramatically reduced values of islet and beta cell count and glucose homeostasis function which neared those measured in single Pttg1 KO mice and were significantly lower than those observed in individual RasGrf1 KO mice. CONCLUSIONS The specific transcriptional profile and signaling behavior of RasgGrf1 KO pancreatic islets, together with the dominance of Pttg1 over RasGrf1 with regards to the generation of these phenotypes in mouse pancreas, suggest that RasGrf1 is an important upstream component of signal transduction pathways regulating Pttg1 expression and controlling beta cell development and physiological responses.
Collapse
Affiliation(s)
| | | | | | | | - Alberto Fernandez-Medarde
- Centro de Investigación del Cáncer, IBMCC (CSIC-USAL), University of Salamanca, Campus Unamuno, 37007 Salamanca, Spain.
| | | |
Collapse
|