1
|
Fenske RJ, Wienkes HN, Peter DC, Schaid MD, Hurley LD, Pennati A, Galipeau J, Kimple ME. Gα z-independent and -dependent Improvements With EPA Supplementation on the Early Type 1 Diabetes Phenotype of NOD Mice. J Endocr Soc 2024; 8:bvae100. [PMID: 38831864 PMCID: PMC11146416 DOI: 10.1210/jendso/bvae100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Indexed: 06/05/2024] Open
Abstract
Prostaglandin E2 (PGE2) is a key mediator of inflammation and is derived from the omega-6 polyunsaturated fatty acid, arachidonic acid (AA). In the β-cell, the PGE2 receptor, Prostaglandin EP3 receptor (EP3), is coupled to the unique heterotrimeric G protein alpha subunit, Gɑz to reduce the production of cyclic adenosine monophosphate (cAMP), a key signaling molecule that activates β-cell function, proliferation, and survival pathways. Nonobese diabetic (NOD) mice are a strong model of type 1 diabetes (T1D), and NOD mice lacking Gɑz are protected from hyperglycemia. Therefore, limiting systemic PGE2 production could potentially improve both the inflammatory and β-cell dysfunction phenotype of T1D. Here, we sought to evaluate the effect of eicosapentaenoic acid (EPA) feeding, which limits PGE2 production, on the early T1D phenotype of NOD mice in the presence and absence of Gαz. Wild-type and Gαz knockout NOD mice were fed a control or EPA-enriched diet for 12 weeks, beginning at age 4 to 5 weeks. Oral glucose tolerance, splenic T-cell populations, islet cytokine/chemokine gene expression, islet insulitis, measurements of β-cell mass, and measurements of β-cell function were quantified. EPA diet feeding and Gɑz loss independently improved different aspects of the early NOD T1D phenotype and coordinated to alter the expression of certain cytokine/chemokine genes and enhance incretin-potentiated insulin secretion. Our results shed critical light on the Gαz-dependent and -independent effects of dietary EPA enrichment and provide a rationale for future research into novel pharmacological and dietary adjuvant therapies for T1D.
Collapse
Affiliation(s)
- Rachel J Fenske
- Research Service, William S. Middleton Memorial VA Hospital, Madison, WI 53705, USA
- Department of Nutritional Sciences, University of Wisconsin–Madison, Madison, WI 53706, USA
- Clinical Research Unit, University of Wisconsin Hospitals and Clinics, Madison, WI 53792, USA
| | - Haley N Wienkes
- Research Service, William S. Middleton Memorial VA Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Darby C Peter
- Research Service, William S. Middleton Memorial VA Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Michael D Schaid
- Research Service, William S. Middleton Memorial VA Hospital, Madison, WI 53705, USA
- Department of Nutritional Sciences, University of Wisconsin–Madison, Madison, WI 53706, USA
- Department of Medicine, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Liam D Hurley
- Research Service, William S. Middleton Memorial VA Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Andrea Pennati
- Department of Medicine, University of Wisconsin–Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Jacques Galipeau
- Department of Medicine, University of Wisconsin–Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Michelle E Kimple
- Research Service, William S. Middleton Memorial VA Hospital, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin–Madison, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin–Madison, Madison, WI 53705, USA
| |
Collapse
|
2
|
Neuman JC, Reuter A, Carbajal KA, Schaid MD, Kelly G, Connors K, Kaiser C, Krause J, Hurley LD, Olvera A, Davis DB, Wisinski JA, Gannon M, Kimple ME. The prostaglandin E 2 EP3 receptor has disparate effects on islet insulin secretion and content in β-cells in a high-fat diet-induced mouse model of obesity. Am J Physiol Endocrinol Metab 2024; 326:E567-E576. [PMID: 38477664 PMCID: PMC11376488 DOI: 10.1152/ajpendo.00061.2023] [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: 02/23/2023] [Revised: 02/07/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024]
Abstract
Signaling through prostaglandin E2 EP3 receptor (EP3) actively contributes to the β-cell dysfunction of type 2 diabetes (T2D). In T2D models, full-body EP3 knockout mice have a significantly worse metabolic phenotype than wild-type controls due to hyperphagia and severe insulin resistance resulting from loss of EP3 in extra-pancreatic tissues, masking any potential beneficial effects of EP3 loss in the β cell. We hypothesized β-cell-specific EP3 knockout (EP3 βKO) mice would be protected from high-fat diet (HFD)-induced glucose intolerance, phenocopying mice lacking the EP3 effector, Gαz, which is much more limited in its tissue distribution. When fed a HFD for 16 wk, though, EP3 βKO mice were partially, but not fully, protected from glucose intolerance. In addition, exendin-4, an analog of the incretin hormone, glucagon-like peptide 1, more strongly potentiated glucose-stimulated insulin secretion in islets from both control diet- and HFD-fed EP3 βKO mice as compared with wild-type controls, with no effect of β-cell-specific EP3 loss on islet insulin content or markers of replication and survival. However, after 26 wk of diet feeding, islets from both control diet- and HFD-fed EP3 βKO mice secreted significantly less insulin as a percent of content in response to stimulatory glucose, with or without exendin-4, with elevated total insulin content unrelated to markers of β-cell replication and survival, revealing severe β-cell dysfunction. Our results suggest that EP3 serves a critical role in temporally regulating β-cell function along the progression to T2D and that there exist Gαz-independent mechanisms behind its effects.NEW & NOTEWORTHY The EP3 receptor is a strong inhibitor of β-cell function and replication, suggesting it as a potential therapeutic target for the disease. Yet, EP3 has protective roles in extrapancreatic tissues. To address this, we designed β-cell-specific EP3 knockout mice and subjected them to high-fat diet feeding to induce glucose intolerance. The negative metabolic phenotype of full-body knockout mice was ablated, and EP3 loss improved glucose tolerance, with converse effects on islet insulin secretion and content.
Collapse
Affiliation(s)
- Joshua C Neuman
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Austin Reuter
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Kathryn A Carbajal
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Michael D Schaid
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Grant Kelly
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Kelsey Connors
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Cecilia Kaiser
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Joshua Krause
- Department of Biology, University of Wisconsin-Lacrosse, La Crosse, Wisconsin, United States
| | - Liam D Hurley
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Angela Olvera
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Dawn Belt Davis
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Jaclyn A Wisinski
- Department of Biology, University of Wisconsin-Lacrosse, La Crosse, Wisconsin, United States
| | - Maureen Gannon
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Wisconsin, United States
| | - Michelle E Kimple
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, United States
| |
Collapse
|
3
|
de Baat A, Meier DT, Rachid L, Fontana A, Böni-Schnetzler M, Donath MY. Cystine/glutamate antiporter System x c- deficiency impairs insulin secretion in mice. Diabetologia 2023; 66:2062-2074. [PMID: 37650924 PMCID: PMC10541846 DOI: 10.1007/s00125-023-05993-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/16/2023] [Indexed: 09/01/2023]
Abstract
AIMS/HYPOTHESIS Glutamate-induced cytotoxicity (excitotoxicity) has been detected in pancreatic beta cells. The cystine/glutamate antiporter System xc- exports glutamate to the extracellular space and is therefore implicated as driving excitotoxicity. As of yet, it has not been investigated whether System xc- contributes to pancreatic islet function. METHODS This study describes the implications of deficiency of System xc- on glucose metabolism in both constitutive and myeloid cell-specific knockout mice using metabolic tests and diet-induced obesity. Pancreatic islets were isolated and analysed for beta cell function, glutathione levels and ER stress. RESULTS Constitutive System xc- deficiency led to an approximately threefold decrease in glutathione levels in the pancreatic islets as well as cystine shortage characterised by upregulation of Chac1. This shortage further manifested as downregulation of beta cell identity genes and a tonic increase in endoplasmic reticulum stress markers, which resulted in diminished insulin secretion both in vitro and in vivo. Myeloid-specific deletion did not have a significant impact on metabolism or islet function. CONCLUSIONS/INTERPRETATION These findings suggest that System xc- is required for glutathione maintenance and insulin production in beta cells and that the system is dispensable for islet macrophage function.
Collapse
Affiliation(s)
- Axel de Baat
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.
- Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - Daniel T Meier
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Leila Rachid
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Adriano Fontana
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marianne Böni-Schnetzler
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marc Y Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
4
|
Wang Y, Regeenes R, Memon M, Rocheleau JV. Insulin C-peptide secretion on-a-chip to measure the dynamics of secretion and metabolism from individual islets. CELL REPORTS METHODS 2023; 3:100602. [PMID: 37820726 PMCID: PMC10626205 DOI: 10.1016/j.crmeth.2023.100602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/16/2023] [Accepted: 09/15/2023] [Indexed: 10/13/2023]
Abstract
First-phase glucose-stimulated insulin secretion is mechanistically linked to type 2 diabetes, yet the underlying metabolism is difficult to discern due to significant islet-to-islet variability. Here, we miniaturize a fluorescence anisotropy immunoassay onto a microfluidic device to measure C-peptide secretion from individual islets as a surrogate for insulin (InsC-chip). This method measures secretion from up to four islets at a time with ∼7 s resolution while providing an optical window for real-time live-cell imaging. Using the InsC-chip, we reveal two glucose-dependent peaks of insulin secretion (i.e., a double peak) within the classically defined 1st phase (<10 min). By combining real-time secretion and live-cell imaging, we show islets transition from glycolytic to oxidative phosphorylation (OxPhos)-driven metabolism at the nadir of the peaks. Overall, these data validate the InsC-chip to measure glucose-stimulated insulin secretion while revealing new dynamics in secretion defined by a shift in glucose metabolism.
Collapse
Affiliation(s)
- Yufeng Wang
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON M5G 1L7, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Romario Regeenes
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON M5G 1L7, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Mahnoor Memon
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON M5G 1L7, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Jonathan V Rocheleau
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON M5G 1L7, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Departments of Medicine and Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
5
|
Krueger ES, Griffin LE, Beales JL, Lloyd TS, Brown NJ, Elison WS, Kay CD, Neilson AP, Tessem JS. Bioavailable Microbial Metabolites of Flavanols Demonstrate Highly Individualized Bioactivity on In Vitro β-Cell Functions Critical for Metabolic Health. Metabolites 2023; 13:801. [PMID: 37512508 PMCID: PMC10385630 DOI: 10.3390/metabo13070801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Dietary flavanols are known for disease preventative properties but are often poorly absorbed. Gut microbiome flavanol metabolites are more bioavailable and may exert protective activities. Using metabolite mixtures extracted from the urine of rats supplemented with flavanols and treated with or without antibiotics, we investigated their effects on INS-1 832/13 β-cell glucose stimulated insulin secretion (GSIS) capacity. We measured insulin secretion under non-stimulatory (low) and stimulatory (high) glucose levels, insulin secretion fold induction, and total insulin content. We conducted treatment-level comparisons, individual-level dose responses, and a responder vs. non-responder predictive analysis of metabolite composition. While the first two analyses did not elucidate treatment effects, metabolites from 9 of the 28 animals demonstrated significant dose responses, regardless of treatment. Differentiation of responders vs. non-responder revealed that levels of native flavanols and valerolactones approached significance for predicting enhanced GSIS, regardless of treatment. Although treatment-level patterns were not discernable, we conclude that the high inter-individual variability shows that metabolite bioactivity on GSIS capacity is less related to flavanol supplementation or antibiotic treatment and may be more associated with the unique microbiome or metabolome of each animal. These findings suggest flavanol metabolite activities are individualized and point to the need for personalized nutrition practices.
Collapse
Affiliation(s)
- Emily S. Krueger
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, UT 84602, USA; (E.S.K.); (J.L.B.); (T.S.L.); (N.J.B.); (W.S.E.)
| | - Laura E. Griffin
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC 28081, USA; (L.E.G.); (C.D.K.); (A.P.N.)
| | - Joseph L. Beales
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, UT 84602, USA; (E.S.K.); (J.L.B.); (T.S.L.); (N.J.B.); (W.S.E.)
| | - Trevor S. Lloyd
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, UT 84602, USA; (E.S.K.); (J.L.B.); (T.S.L.); (N.J.B.); (W.S.E.)
| | - Nathan J. Brown
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, UT 84602, USA; (E.S.K.); (J.L.B.); (T.S.L.); (N.J.B.); (W.S.E.)
| | - Weston S. Elison
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, UT 84602, USA; (E.S.K.); (J.L.B.); (T.S.L.); (N.J.B.); (W.S.E.)
| | - Colin D. Kay
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC 28081, USA; (L.E.G.); (C.D.K.); (A.P.N.)
| | - Andrew P. Neilson
- Plants for Human Health Institute, Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC 28081, USA; (L.E.G.); (C.D.K.); (A.P.N.)
| | - Jeffery S. Tessem
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, UT 84602, USA; (E.S.K.); (J.L.B.); (T.S.L.); (N.J.B.); (W.S.E.)
| |
Collapse
|
6
|
Brownrigg GP, Xia YH, Chu CMJ, Wang S, Chao C, Zhang JA, Skovsø S, Panzhinskiy E, Hu X, Johnson JD, Rideout EJ. Sex differences in islet stress responses support female β cell resilience. Mol Metab 2023; 69:101678. [PMID: 36690328 PMCID: PMC9971554 DOI: 10.1016/j.molmet.2023.101678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/07/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE Pancreatic β cells play a key role in maintaining glucose homeostasis; dysfunction of this critical cell type causes type 2 diabetes (T2D). Emerging evidence points to sex differences in β cells, but few studies have examined male-female differences in β cell stress responses and resilience across multiple contexts, including diabetes. Here, we address the need for high-quality information on sex differences in β cell and islet gene expression and function using both human and rodent samples. METHODS In humans, we compared β cell gene expression and insulin secretion in donors with T2D to non-diabetic donors in both males and females. In mice, we generated a well-powered islet RNAseq dataset from 20-week-old male and female siblings with similar insulin sensitivity. Our unbiased gene expression analysis pointed to a sex difference in the endoplasmic reticulum (ER) stress response. Based on this analysis, we hypothesized female islets would be more resilient to ER stress than male islets. To test this, we subjected islets isolated from age-matched male and female mice to thapsigargin treatment and monitored protein synthesis, cell death, and β cell insulin production and secretion. Transcriptomic and proteomic analyses were used to characterize sex differences in islet responses to ER stress. RESULTS Our single-cell analysis of human β cells revealed sex-specific changes to gene expression and function in T2D, correlating with more robust insulin secretion in human islets isolated from female donors with T2D compared to male donors with T2D. In mice, RNA sequencing revealed differential enrichment of unfolded protein response pathway-associated genes, where female islets showed higher expression of genes linked with protein synthesis, folding, and processing. This differential expression was physiologically significant, as islets isolated from female mice were more resilient to ER stress induction with thapsigargin. Specifically, female islets showed a greater ability to maintain glucose-stimulated insulin production and secretion during ER stress compared with males. CONCLUSIONS Our data demonstrate sex differences in β cell gene expression in both humans and mice, and that female β cells show a greater ability to maintain glucose-stimulated insulin secretion across multiple physiological and pathological contexts.
Collapse
Affiliation(s)
- George P Brownrigg
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yi Han Xia
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Chieh Min Jamie Chu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Su Wang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Charlotte Chao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Jiashuo Aaron Zhang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Søs Skovsø
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Evgeniy Panzhinskiy
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Xiaoke Hu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - James D Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
7
|
Cujba AM, Alvarez-Fallas ME, Pedraza-Arevalo S, Laddach A, Shepherd MH, Hattersley AT, Watt FM, Sancho R. An HNF1α truncation associated with maturity-onset diabetes of the young impairs pancreatic progenitor differentiation by antagonizing HNF1β function. Cell Rep 2022; 38:110425. [PMID: 35235779 PMCID: PMC8905088 DOI: 10.1016/j.celrep.2022.110425] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/23/2021] [Accepted: 02/02/2022] [Indexed: 01/16/2023] Open
Abstract
The HNF1αp291fsinsC truncation is the most common mutation associated with maturity-onset diabetes of the young 3 (MODY3). Although shown to impair HNF1α signaling, the mechanism by which HNF1αp291fsinsC causes MODY3 is not fully understood. Here we use MODY3 patient and CRISPR/Cas9-engineered human induced pluripotent stem cells (hiPSCs) grown as 3D organoids to investigate how HNF1αp291fsinsC affects hiPSC differentiation during pancreatic development. HNF1αp291fsinsC hiPSCs shows reduced pancreatic progenitor and β cell differentiation. Mechanistically, HNF1αp291fsinsC interacts with HNF1β and inhibits its function, and disrupting this interaction partially rescues HNF1β-dependent transcription. HNF1β overexpression in the HNF1αp291fsinsC patient organoid line increases PDX1+ progenitors, while HNF1β overexpression in the HNF1αp291fsinsC patient iPSC line partially rescues β cell differentiation. Our study highlights the capability of pancreas progenitor-derived organoids to model disease in vitro. Additionally, it uncovers an HNF1β-mediated mechanism linked to HNF1α truncation that affects progenitor differentiation and could explain the clinical heterogeneity observed in MODY3 patients. MODY3 patient and CRISPR/Cas9 HNF1αp291fsinsC mutated iPSC lines are generated Mutant iPSCs show deficient pancreatic progenitor and β cell differentiation Mutant truncated HNF1α protein binds wild-type HNF1β protein to hinder its function HNF1β overexpression in MODY3 iPSC line partially rescues β cell differentiation
Collapse
Affiliation(s)
- Ana-Maria Cujba
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | | | | | | | | | | | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Rocio Sancho
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK; Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
8
|
Gut Metabolite Trimethylamine N-Oxide Protects INS-1 β-Cell and Rat Islet Function under Diabetic Glucolipotoxic Conditions. Biomolecules 2021; 11:biom11121892. [PMID: 34944536 PMCID: PMC8699500 DOI: 10.3390/biom11121892] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
Serum accumulation of the gut microbial metabolite trimethylamine N-oxide (TMAO) is associated with high caloric intake and type 2 diabetes (T2D). Impaired pancreatic β-cell function is a hallmark of diet-induced T2D, which is linked to hyperglycemia and hyperlipidemia. While TMAO production via the gut microbiome-liver axis is well defined, its molecular effects on metabolic tissues are unclear, since studies in various tissues show deleterious and beneficial TMAO effects. We investigated the molecular effects of TMAO on functional β-cell mass. We hypothesized that TMAO may damage functional β-cell mass by inhibiting β-cell viability, survival, proliferation, or function to promote T2D pathogenesis. We treated INS-1 832/13 β-cells and primary rat islets with physiological TMAO concentrations and compared functional β-cell mass under healthy standard cell culture (SCC) and T2D-like glucolipotoxic (GLT) conditions. GLT significantly impeded β-cell mass and function by inducing oxidative and endoplasmic reticulum (ER) stress. TMAO normalized GLT-mediated damage in β-cells and primary islet function. Acute 40µM TMAO recovered insulin production, insulin granule formation, and insulin secretion by upregulating the IRE1α unfolded protein response to GLT-induced ER and oxidative stress. These novel results demonstrate that TMAO protects β-cell function and suggest that TMAO may play a beneficial molecular role in diet-induced T2D conditions.
Collapse
|
9
|
Wisinski JA, Reuter A, Peter DC, Schaid MD, Fenske RJ, Kimple ME. Prostaglandin EP3 receptor signaling is required to prevent insulin hypersecretion and metabolic dysfunction in a non-obese mouse model of insulin resistance. Am J Physiol Endocrinol Metab 2021; 321:E479-E489. [PMID: 34229444 PMCID: PMC8560379 DOI: 10.1152/ajpendo.00051.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
When homozygous for the LeptinOb mutation (Ob), Black-and-Tan Brachyury (BTBR) mice become morbidly obese and severely insulin resistant, and by 10 wk of age, frankly diabetic. Previous work has shown prostaglandin EP3 receptor (EP3) expression and activity is upregulated in islets from BTBR-Ob mice as compared with lean controls, actively contributing to their β-cell dysfunction. In this work, we aimed to test the impact of β-cell-specific EP3 loss on the BTBR-Ob phenotype by crossing Ptger3 floxed mice with the rat insulin promoter (RIP)-CreHerr driver strain. Instead, germline recombination of the floxed allele in the founder mouse-an event whose prevalence we identified as directly associated with underlying insulin resistance of the background strain-generated a full-body knockout. Full-body EP3 loss provided no diabetes protection to BTBR-Ob mice but, unexpectedly, significantly worsened BTBR-lean insulin resistance and glucose tolerance. This in vivo phenotype was not associated with changes in β-cell fractional area or markers of β-cell replication ex vivo. Instead, EP3-null BTBR-lean islets had essentially uncontrolled insulin hypersecretion. The selective upregulation of constitutively active EP3 splice variants in islets from young, lean BTBR mice as compared with C57BL/6J, where no phenotype of EP3 loss has been observed, provides a potential explanation for the hypersecretion phenotype. In support of this, high islet EP3 expression in Balb/c females versus Balb/c males was fully consistent with their sexually dimorphic metabolic phenotype after loss of EP3-coupled Gαz protein. Taken together, our findings provide a new dimension to the understanding of EP3 as a critical brake on insulin secretion.NEW & NOTEWORTHY Islet prostaglandin EP3 receptor (EP3) signaling is well known as upregulated in the pathophysiological conditions of type 2 diabetes, contributing to β-cell dysfunction. Unexpected findings in mouse models of non-obese insulin sensitivity and resistance provide a new dimension to our understanding of EP3 as a key modulator of insulin secretion. A previously unknown relationship between mouse insulin resistance and the penetrance of rat insulin promoter-driven germline floxed allele recombination is critical to consider when creating β-cell-specific knockouts.
Collapse
Affiliation(s)
- Jaclyn A Wisinski
- Department of Biology, University of Wisconsin-LaCrosse, La Crosse, Wisconsin
| | - Austin Reuter
- Research Service, William S. Middleton Memorial VA Hospital, Madison, Wisconsin
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
| | - Darby C Peter
- Research Service, William S. Middleton Memorial VA Hospital, Madison, Wisconsin
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
| | - Michael D Schaid
- Research Service, William S. Middleton Memorial VA Hospital, Madison, Wisconsin
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Rachel J Fenske
- Research Service, William S. Middleton Memorial VA Hospital, Madison, Wisconsin
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
| | - Michelle E Kimple
- Research Service, William S. Middleton Memorial VA Hospital, Madison, Wisconsin
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
10
|
Truchan NA, Fenske RJ, Sandhu HK, Weeks AM, Patibandla C, Wancewicz B, Pabich S, Reuter A, Harrington JM, Brill AL, Peter DC, Nall R, Daniels M, Punt M, Kaiser CE, Cox ED, Ge Y, Davis DB, Kimple ME. Human Islet Expression Levels of Prostaglandin E 2 Synthetic Enzymes, But Not Prostaglandin EP3 Receptor, Are Positively Correlated with Markers of β-Cell Function and Mass in Nondiabetic Obesity. ACS Pharmacol Transl Sci 2021; 4:1338-1348. [PMID: 34423270 DOI: 10.1021/acsptsci.1c00045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Indexed: 01/06/2023]
Abstract
Elevated islet production of prostaglandin E2 (PGE2), an arachidonic acid metabolite, and expression of prostaglandin E2 receptor subtype EP3 (EP3) are well-known contributors to the β-cell dysfunction of type 2 diabetes (T2D). Yet, many of the same pathophysiological conditions exist in obesity, and little is known about how the PGE2 production and signaling pathway influences nondiabetic β-cell function. In this work, plasma arachidonic acid and PGE2 metabolite levels were quantified in a cohort of nondiabetic and T2D human subjects to identify their relationship with glycemic control, obesity, and systemic inflammation. In order to link these findings to processes happening at the islet level, cadaveric human islets were subject to gene expression and functional assays. Interleukin-6 (IL-6) and cyclooxygenase-2 (COX-2) mRNA levels, but not those of EP3, positively correlated with donor body mass index (BMI). IL-6 expression also strongly correlated with the expression of COX-2 and other PGE2 synthetic pathway genes. Insulin secretion assays using an EP3-specific antagonist confirmed functionally relevant upregulation of PGE2 production. Yet, islets from obese donors were not dysfunctional, secreting just as much insulin in basal and stimulatory conditions as those from nonobese donors as a percent of content. Islet insulin content, on the other hand, was increased with both donor BMI and islet COX-2 expression, while EP3 expression was unaffected. We conclude that upregulated islet PGE2 production may be part of the β-cell adaption response to obesity and insulin resistance that only becomes dysfunctional when both ligand and receptor are highly expressed in T2D.
Collapse
Affiliation(s)
- Nathan A Truchan
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States
| | - Rachel J Fenske
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States.,Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Harpreet K Sandhu
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States
| | - Alicia M Weeks
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States
| | - Chinmai Patibandla
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States
| | - Benjamin Wancewicz
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Samantha Pabich
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Austin Reuter
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States
| | - Jeffrey M Harrington
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States
| | - Allison L Brill
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States
| | - Darby C Peter
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States
| | - Randall Nall
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States
| | - Michael Daniels
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States
| | - Margaret Punt
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Cecilia E Kaiser
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States
| | - Elizabeth D Cox
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin 53792, United States
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Dawn B Davis
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States.,Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Michelle E Kimple
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States.,Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, United States.,Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States.,Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
11
|
A Practical Guide to Rodent Islet Isolation and Assessment Revisited. Biol Proced Online 2021; 23:7. [PMID: 33641671 PMCID: PMC7919091 DOI: 10.1186/s12575-021-00143-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Insufficient insulin secretion is a key component of both type 1 and type 2 diabetes. Since insulin is released by the islets of Langerhans, obtaining viable and functional islets is critical for research and transplantation. The effective and efficient isolation of these small islands of endocrine cells from the sea of exocrine tissue that is the rest of the pancreas is not necessarily simple or quick. Choosing and administering the digestive enzyme, separation of the islets from acinar tissue, and culture of islets are all things that must be considered. The purpose of this review is to provide a history of the development of islet isolation procedures and to serve as a practical guide to rodent islet research for newcomers to islet biology. We discuss key elements of mouse islet isolation including choosing collagenase, the digestion process, purification of islets using a density gradient, and islet culture conditions. In addition, this paper reviews techniques for assessing islet viability and function such as visual assessment, glucose-stimulated insulin secretion and intracellular calcium measurements. A detailed protocol is provided that describes a common method our laboratory uses to obtain viable and functional mouse islets for in vitro study. This review thus provides a strong foundation for successful procurement and purification of high-quality mouse islets for research purposes.
Collapse
|
12
|
Schaid MD, Zhu Y, Richardson NE, Patibandla C, Ong IM, Fenske RJ, Neuman JC, Guthery E, Reuter A, Sandhu HK, Fuller MH, Cox ED, Davis DB, Layden BT, Brasier AR, Lamming DW, Ge Y, Kimple ME. Systemic Metabolic Alterations Correlate with Islet-Level Prostaglandin E 2 Production and Signaling Mechanisms That Predict β-Cell Dysfunction in a Mouse Model of Type 2 Diabetes. Metabolites 2021; 11:metabo11010058. [PMID: 33467110 PMCID: PMC7830513 DOI: 10.3390/metabo11010058] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 12/18/2022] Open
Abstract
The transition from β-cell compensation to β-cell failure is not well understood. Previous works by our group and others have demonstrated a role for Prostaglandin EP3 receptor (EP3), encoded by the Ptger3 gene, in the loss of functional β-cell mass in Type 2 diabetes (T2D). The primary endogenous EP3 ligand is the arachidonic acid metabolite prostaglandin E2 (PGE2). Expression of the pancreatic islet EP3 and PGE2 synthetic enzymes and/or PGE2 excretion itself have all been shown to be upregulated in primary mouse and human islets isolated from animals or human organ donors with established T2D compared to nondiabetic controls. In this study, we took advantage of a rare and fleeting phenotype in which a subset of Black and Tan BRachyury (BTBR) mice homozygous for the Leptinob/ob mutation—a strong genetic model of T2D—were entirely protected from fasting hyperglycemia even with equal obesity and insulin resistance as their hyperglycemic littermates. Utilizing this model, we found numerous alterations in full-body metabolic parameters in T2D-protected mice (e.g., gut microbiome composition, circulating pancreatic and incretin hormones, and markers of systemic inflammation) that correlate with improvements in EP3-mediated β-cell dysfunction.
Collapse
Affiliation(s)
- Michael D. Schaid
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.J.F.); (J.C.N.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Yanlong Zhu
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; (Y.Z.); (Y.G.)
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Nicole E. Richardson
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Chinmai Patibandla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Irene M. Ong
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, USA;
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Rachel J. Fenske
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.J.F.); (J.C.N.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Joshua C. Neuman
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.J.F.); (J.C.N.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Erin Guthery
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Austin Reuter
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Harpreet K. Sandhu
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Miles H. Fuller
- Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL 60612, USA; (M.H.F.); (B.T.L.)
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| | - Elizabeth D. Cox
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53792, USA;
| | - Dawn B. Davis
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.J.F.); (J.C.N.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Brian T. Layden
- Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL 60612, USA; (M.H.F.); (B.T.L.)
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| | - Allan R. Brasier
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Institute for Clinical and Translational Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.J.F.); (J.C.N.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; (Y.Z.); (Y.G.)
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michelle E. Kimple
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.J.F.); (J.C.N.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; (Y.Z.); (Y.G.)
- Correspondence: ; Tel.: +1-1-608-265-5627
| |
Collapse
|
13
|
Schaid MD, Green CL, Peter DC, Gallagher SJ, Guthery E, Carbajal KA, Harrington JM, Kelly GM, Reuter A, Wehner ML, Brill AL, Neuman JC, Lamming DW, Kimple ME. Agonist-independent Gα z activity negatively regulates beta-cell compensation in a diet-induced obesity model of type 2 diabetes. J Biol Chem 2020; 296:100056. [PMID: 33172888 PMCID: PMC7948463 DOI: 10.1074/jbc.ra120.015585] [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: 08/10/2020] [Revised: 11/04/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022] Open
Abstract
The inhibitory G protein alpha-subunit (Gαz) is an important modulator of beta-cell function. Full-body Gαz-null mice are protected from hyperglycemia and glucose intolerance after long-term high-fat diet (HFD) feeding. In this study, at a time point in the feeding regimen where WT mice are only mildly glucose intolerant, transcriptomics analyses reveal islets from HFD-fed Gαz KO mice have a dramatically altered gene expression pattern as compared with WT HFD-fed mice, with entire gene pathways not only being more strongly upregulated or downregulated versus control-diet fed groups but actually reversed in direction. Genes involved in the “pancreatic secretion” pathway are the most strongly differentially regulated: a finding that correlates with enhanced islet insulin secretion and decreased glucagon secretion at the study end. The protection of Gαz-null mice from HFD-induced diabetes is beta-cell autonomous, as beta cell–specific Gαz-null mice phenocopy the full-body KOs. The glucose-stimulated and incretin-potentiated insulin secretion response of islets from HFD-fed beta cell–specific Gαz-null mice is significantly improved as compared with islets from HFD-fed WT controls, which, along with no impact of Gαz loss or HFD feeding on beta-cell proliferation or surrogates of beta-cell mass, supports a secretion-specific mechanism. Gαz is coupled to the prostaglandin EP3 receptor in pancreatic beta cells. We confirm the EP3γ splice variant has both constitutive and agonist-sensitive activity to inhibit cAMP production and downstream beta-cell function, with both activities being dependent on the presence of beta-cell Gαz.
Collapse
Affiliation(s)
- Michael D Schaid
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Interdepartmental Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Cara L Green
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Darby C Peter
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Shannon J Gallagher
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Erin Guthery
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Kathryn A Carbajal
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Jeffrey M Harrington
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Grant M Kelly
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Austin Reuter
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Molly L Wehner
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Allison L Brill
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Joshua C Neuman
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Interdepartmental Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Dudley W Lamming
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Interdepartmental Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Michelle E Kimple
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA; Interdepartmental Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA; Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA; Department of Cell and Regenerative Biology, University of Wisconsin- Madison School of Medicine and Public Health, Madison, Wisconsin, USA.
| |
Collapse
|
14
|
Li WH. Functional analysis of islet cells in vitro, in situ, and in vivo. Semin Cell Dev Biol 2020; 103:14-19. [PMID: 32081627 DOI: 10.1016/j.semcdb.2020.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/05/2020] [Indexed: 12/20/2022]
Abstract
The islet of Langerhans contains at least five types of endocrine cells producing distinct hormones. In response to nutrient or neuronal stimulation, islet endocrine cells release biochemicals including peptide hormones to regulate metabolism and to control glucose homeostasis. It is now recognized that malfunction of islet cells, notably insufficient insulin release of β-cells and hypersecretion of glucagon from α-cells, represents a causal event leading to hyperglycemia and frank diabetes, a disease that is increasing at an alarming rate to reach an epidemic level worldwide. Understanding the mechanisms regulating stimulus-secretion coupling and investigating how islet β-cells maintain a robust secretory activity are important topics in islet biology and diabetes research. To facilitate such studies, a number of biological systems and assay platforms have been developed for the functional analysis of islet cells. These technologies have enabled detailed analyses of individual islets at the cellular level, either in vitro, in situ, or in vivo.
Collapse
Affiliation(s)
- Wen-Hong Li
- Departments of Cell Biology and of Biochemistry, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX, 75390-9039, United States.
| |
Collapse
|
15
|
Taddeo EP, Alsabeeh N, Baghdasarian S, Wikstrom JD, Ritou E, Sereda S, Erion K, Li J, Stiles L, Abdulla M, Swanson Z, Wilhelm JJ, Bellin MD, Kibbey RG, Liesa M, Shirihai OS. Mitochondrial Proton Leak Regulated by Cyclophilin D Elevates Insulin Secretion in Islets at Nonstimulatory Glucose Levels. Diabetes 2020; 69:131-145. [PMID: 31740442 PMCID: PMC6971491 DOI: 10.2337/db19-0379] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022]
Abstract
Fasting hyperinsulinemia precedes the development of type 2 diabetes. However, it is unclear whether fasting insulin hypersecretion is a primary driver of insulin resistance or a consequence of the progressive increase in fasting glycemia induced by insulin resistance in the prediabetic state. Herein, we have discovered a mechanism that specifically regulates non-glucose-stimulated insulin secretion (NGSIS) in pancreatic islets that is activated by nonesterified free fatty acids, the major fuel used by β-cells during fasting. We show that the mitochondrial permeability transition pore regulator cyclophilin D (CypD) promotes NGSIS, but not glucose-stimulated insulin secretion, by increasing mitochondrial proton leak. Islets from prediabetic obese mice show significantly higher CypD-dependent proton leak and NGSIS compared with lean mice. Proton leak-mediated NGSIS is conserved in human islets and is stimulated by exposure to nonesterified free fatty acids at concentrations observed in obese subjects. Mechanistically, proton leak activates islet NGSIS independently of mitochondrial ATP synthesis but ultimately requires closure of the KATP channel. In summary, we have described a novel nonesterified free fatty acid-stimulated pathway that selectively drives pancreatic islet NGSIS, which may be therapeutically exploited as an alternative way to halt fasting hyperinsulinemia and the progression of type 2 diabetes.
Collapse
Affiliation(s)
- Evan P Taddeo
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Nour Alsabeeh
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Department of Physiology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Siyouneh Baghdasarian
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Jakob D Wikstrom
- Dermatology and Venereology Unit, Department of Medicine, Karolinska Institutet, and Department of Dermato-Venereology, Karolinska University Hospital, Stockholm, Sweden
| | - Eleni Ritou
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Samuel Sereda
- Endocrinology, Diabetes, Nutrition and Weight Management Section, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Karel Erion
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Jin Li
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Linsey Stiles
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Muhamad Abdulla
- Department of Surgery and Schulze Diabetes Institute, University of Minnesota School of Medicine, Minneapolis, MN
| | - Zachary Swanson
- Department of Surgery and Schulze Diabetes Institute, University of Minnesota School of Medicine, Minneapolis, MN
| | - Joshua J Wilhelm
- Department of Surgery and Schulze Diabetes Institute, University of Minnesota School of Medicine, Minneapolis, MN
| | - Melena D Bellin
- Department of Surgery and Schulze Diabetes Institute, University of Minnesota School of Medicine, Minneapolis, MN
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN
| | - Richard G Kibbey
- Departments of Internal Medicine (Endocrinology) and Cellular & Molecular Physiology, Yale University, New Haven, CT
| | - Marc Liesa
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| | - Orian S Shirihai
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
16
|
Chung H, Hong SJ, Choi SW, Park CG. The effect of preexisting HMGB1 within fetal bovine serum on murine pancreatic beta cell biology. Islets 2020; 12:1-8. [PMID: 31935155 PMCID: PMC7064295 DOI: 10.1080/19382014.2019.1696128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
High-mobility group box 1 (HMGB1) can act as a structural protein of the chromatin and at the same time as a mediator of the immune system. Its high correlation with the graft acceptance in pancreatic islet recipients makes it a biomarker in islet transplantation. With the suspicion that preexisting HMGB1 in the fetal bovine serum (FBS) would be detrimental to the viability and function of murine beta cells, HMGB1 was removed from FBS and its impact was investigated. Interestingly, the elimination of HMGB1 from FBS seemed unfavorable to the viability and function of cultured murine beta cells, suggesting that the preexisting HMGB1 in the FBS may be an indispensable component of islet cell culture.
Collapse
Affiliation(s)
- Hyunwoo Chung
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Ji Hong
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - So Won Choi
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Chung-Gyu Park
- Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Biomedical Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Institute of Endemic Diseases, Seoul National University College of Medicine, Seoul, Korea
- CONTACT Chung-Gyu Park Department of Microbiology and Immunology, Department of Biomedical Sciences, Xenotransplantation Research Center, Seoul National University College of Medicine, 103 Daehak-ro Jongno-gu, Seoul 110-799, Korea
| |
Collapse
|
17
|
Pratt EPS, Harvey KE, Salyer AE, Hockerman GH. Regulation of cAMP accumulation and activity by distinct phosphodiesterase subtypes in INS-1 cells and human pancreatic β-cells. PLoS One 2019; 14:e0215188. [PMID: 31442224 PMCID: PMC6707593 DOI: 10.1371/journal.pone.0215188] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 08/11/2019] [Indexed: 01/09/2023] Open
Abstract
Pancreatic β-cells express multiple phosphodiesterase (PDE) subtypes, but the specific roles for each in β-cell function, particularly in humans, is not clear. We evaluated the cellular role of PDE1, PDE3, and PDE4 activity in the rat insulinoma cell line INS-1 and in primary human β-cells using subtype-selective PDE inhibitors. Using a genetically encoded, FRET-based cAMP sensor, we found that the PDE1 inhibitor 8MM-IBMX, elevated cAMP levels in the absence of glucose to a greater extent than either the PDE3 inhibitor cilostamide or the PDE4 inhibitor rolipram. In 18 mM glucose, PDE1 inhibition elevated cAMP levels to a greater extent than PDE3 inhibition in INS-1 cells, while PDE4 inhibition was without effect. Inhibition of PDE1 or PDE4, but not PDE3, potentiated glucose-stimulated insulin secretion in INS-1 cells. PDE1 inhibition, but not PDE3 or PDE4 inhibition, reduced palmitate-induced caspase-3/7 activation, and enhanced CREB phosphorylation in INS-1 cells. In human β-cells, only PDE3 or PDE4 inhibition increased cAMP levels in 1.7 mM glucose, but PDE1, PDE3, or PDE4 inhibition potentiated cAMP levels in 16.7 mM glucose. Inhibition of PDE1 or PDE4 increased cAMP levels to a greater extent in 16.7 mM glucose than in 1.7 mM glucose in human β-cells. In contrast, elevation of cAMP levels by PDE3 inhibition was not different at these glucose concentrations. PDE1 inhibition also potentiated insulin secretion from human islets, suggesting that the role of PDE1 may be conserved between INS-1 cells and human pancreatic β-cells. Our results suggest that inhibition of PDE1 may be a useful strategy to potentiate glucose-stimulated insulin secretion, and to protect β-cells from the toxic effects of excess fatty acids.
Collapse
Affiliation(s)
- Evan P. S. Pratt
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States of America
- Purdue University Interdisciplinary Life Science Program, Purdue University, West Lafayette, IN, United States of America
| | - Kyle E. Harvey
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States of America
| | - Amy E. Salyer
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States of America
| | - Gregory H. Hockerman
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States of America
- * E-mail:
| |
Collapse
|
18
|
Yu D, Yang SE, Miller BR, Wisinski JA, Sherman DS, Brinkman JA, Tomasiewicz JL, Cummings NE, Kimple ME, Cryns VL, Lamming DW. Short-term methionine deprivation improves metabolic health via sexually dimorphic, mTORC1-independent mechanisms. FASEB J 2018; 32:3471-3482. [PMID: 29401631 PMCID: PMC5956241 DOI: 10.1096/fj.201701211r] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/22/2018] [Indexed: 12/28/2022]
Abstract
Obesity and diabetes are major challenges to global health, and there is an urgent need for interventions that promote weight loss. Dietary restriction of methionine promotes leanness and improves metabolic health in mice and humans. However, poor long-term adherence to this diet limits its translational potential. In this study, we develop a short-term methionine deprivation (MD) regimen that preferentially reduces fat mass, restoring normal body weight and glycemic control to diet-induced obese mice of both sexes. The benefits of MD do not accrue from calorie restriction, but instead result from increased energy expenditure. MD promotes increased energy expenditure in a sex-specific manner, inducing the fibroblast growth factor (Fgf)-21-uncoupling protein (Ucp)-1 axis only in males. Methionine is an agonist of the protein kinase mechanistic target of rapamycin complex (mTORC)-1, which has been proposed to play a key role in the metabolic response to amino acid-restricted diets. In our study, we used a mouse model of constitutive hepatic mTORC1 activity and demonstrate that suppression of hepatic mTORC1 signaling is not required for the metabolic effects of MD. Our study sheds new light on the mechanisms by which dietary methionine regulates metabolic health and demonstrates the translational potential of MD for the treatment of obesity and type 2 diabetes.-Yu, D., Yang, S. E., Miller, B. R., Wisinski, J. A., Sherman, D. S., Brinkman, J. A., Tomasiewicz, J. L., Cummings, N. E., Kimple, M. E., Cryns, V. L., Lamming, D. W. Short-term methionine deprivation improves metabolic health via sexually dimorphic, mTORC1-independent mechanisms.
Collapse
Affiliation(s)
- Deyang Yu
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
- Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Shany E. Yang
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Blake R. Miller
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Jaclyn A. Wisinski
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
- Department of Biology, University of Wisconsin-La Crosse, La Crosse, Wisconsin, USA
| | - Dawn S. Sherman
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Jacqueline A. Brinkman
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Jay L. Tomasiewicz
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Nicole E. Cummings
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
- Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA; and
| | - Michelle E. Kimple
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
- Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA; and
| | - Vincent L. Cryns
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| | - Dudley W. Lamming
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
- Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA; and
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin, USA
| |
Collapse
|
19
|
Cummings NE, Williams EM, Kasza I, Konon EN, Schaid MD, Schmidt BA, Poudel C, Sherman DS, Yu D, Arriola Apelo SI, Cottrell SE, Geiger G, Barnes ME, Wisinski JA, Fenske RJ, Matkowskyj KA, Kimple ME, Alexander CM, Merrins MJ, Lamming DW. Restoration of metabolic health by decreased consumption of branched-chain amino acids. J Physiol 2017; 596:623-645. [PMID: 29266268 DOI: 10.1113/jp275075] [Citation(s) in RCA: 238] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/20/2017] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS We recently found that feeding healthy mice a diet with reduced levels of branched-chain amino acids (BCAAs), which are associated with insulin resistance in both humans and rodents, modestly improves glucose tolerance and slows fat mass gain. In the present study, we show that a reduced BCAA diet promotes rapid fat mass loss without calorie restriction in obese mice. Selective reduction of dietary BCAAs also restores glucose tolerance and insulin sensitivity to obese mice, even as they continue to consume a high-fat, high-sugar diet. A low BCAA diet transiently induces FGF21 (fibroblast growth factor 21) and increases energy expenditure. We suggest that dietary protein quality (i.e. the precise macronutrient composition of dietary protein) may impact the effectiveness of weight loss diets. ABSTRACT Obesity and diabetes are increasing problems around the world, and although even moderate weight loss can improve metabolic health, reduced calorie diets are notoriously difficult to sustain. Branched-chain amino acids (BCAAs; leucine, isoleucine and valine) are elevated in the blood of obese, insulin-resistant humans and rodents. We recently demonstrated that specifically reducing dietary levels of BCAAs has beneficial effects on the metabolic health of young, growing mice, improving glucose tolerance and modestly slowing fat mass gain. In the present study, we examine the hypothesis that reducing dietary BCAAs will promote weight loss, reduce adiposity, and improve blood glucose control in diet-induced obese mice with pre-existing metabolic syndrome. We find that specifically reducing dietary BCAAs rapidly reverses diet-induced obesity and improves glucoregulatory control in diet-induced obese mice. Most dramatically, mice eating an otherwise unhealthy high-calorie, high-sugar Western diet with reduced levels of BCAAs lost weight and fat mass rapidly until regaining a normal weight. Importantly, this normalization of weight was mediated not by caloric restriction or increased activity, but by increased energy expenditure, and was accompanied by a transient induction of the energy balance regulating hormone FGF21 (fibroblast growth factor 21). Consumption of a Western diet reduced in BCAAs was also accompanied by a dramatic improvement in glucose tolerance and insulin resistance. Our results link dietary BCAAs with the regulation of metabolic health and energy balance in obese animals, and suggest that specifically reducing dietary BCAAs may represent a highly translatable option for the treatment of obesity and insulin resistance.
Collapse
Affiliation(s)
- Nicole E Cummings
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth M Williams
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Ildiko Kasza
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth N Konon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michael D Schaid
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Brian A Schmidt
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chetan Poudel
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Dawn S Sherman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Deyang Yu
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Sebastian I Arriola Apelo
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Department of Dairy Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Sara E Cottrell
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Rural and Urban Scholars in Community Health Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Gabriella Geiger
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Macy E Barnes
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Jaclyn A Wisinski
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Rachel J Fenske
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Kristina A Matkowskyj
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA.,Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle E Kimple
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA.,Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA.,Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Caroline M Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Matthew J Merrins
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA.,Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA.,Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
20
|
Neuman JC, Schaid MD, Brill AL, Fenske RJ, Kibbe CR, Fontaine DA, Sdao SM, Brar HK, Connors KM, Wienkes HN, Eliceiri KW, Merrins MJ, Davis DB, Kimple ME. Enriching Islet Phospholipids With Eicosapentaenoic Acid Reduces Prostaglandin E 2 Signaling and Enhances Diabetic β-Cell Function. Diabetes 2017; 66:1572-1585. [PMID: 28193789 PMCID: PMC5440023 DOI: 10.2337/db16-1362] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 02/09/2017] [Indexed: 12/26/2022]
Abstract
Prostaglandin E2 (PGE2) is derived from arachidonic acid, whereas PGE3 is derived from eicosapentaenoic acid (EPA) using the same downstream metabolic enzymes. Little is known about the impact of EPA and PGE3 on β-cell function, particularly in the diabetic state. In this work, we determined that PGE3 elicits a 10-fold weaker reduction in glucose-stimulated insulin secretion through the EP3 receptor as compared with PGE2 We tested the hypothesis that enriching pancreatic islet cell membranes with EPA, thereby reducing arachidonic acid abundance, would positively impact β-cell function in the diabetic state. EPA-enriched islets isolated from diabetic BTBR Leptinob/ob mice produced significantly less PGE2 and more PGE3 than controls, correlating with improved glucose-stimulated insulin secretion. NAD(P)H fluorescence lifetime imaging showed that EPA acts downstream and independently of mitochondrial function. EPA treatment also reduced islet interleukin-1β expression, a proinflammatory cytokine known to stimulate prostaglandin production and EP3 expression. Finally, EPA feeding improved glucose tolerance and β-cell function in a mouse model of diabetes that incorporates a strong immune phenotype: the NOD mouse. In sum, increasing pancreatic islet EPA abundance improves diabetic β-cell function through both direct and indirect mechanisms that converge on reduced EP3 signaling.
Collapse
Affiliation(s)
- Joshua C Neuman
- Interdisciplinary Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, WI
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Michael D Schaid
- Interdisciplinary Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, WI
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Allison L Brill
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Rachel J Fenske
- Interdisciplinary Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, WI
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Carly R Kibbe
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Danielle A Fontaine
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Sophia M Sdao
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Integrated Program in Biochemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Harpreet K Brar
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Kelsey M Connors
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Haley N Wienkes
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Kevin W Eliceiri
- Department of Biomedical Engineering, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Matthew J Merrins
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
- Integrated Program in Biochemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| | - Dawn B Davis
- Interdisciplinary Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
- Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Michelle E Kimple
- Interdisciplinary Graduate Program in Nutritional Sciences, College of Agriculture and Life Sciences, University of Wisconsin-Madison, Madison, WI
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI
| |
Collapse
|
21
|
Fenske RJ, Cadena MT, Harenda QE, Wienkes HN, Carbajal K, Schaid MD, Laundre E, Brill AL, Truchan NA, Brar H, Wisinski J, Cai J, Graham TE, Engin F, Kimple ME. The Inhibitory G Protein α-Subunit, Gαz, Promotes Type 1 Diabetes-Like Pathophysiology in NOD Mice. Endocrinology 2017; 158:1645-1658. [PMID: 28419211 PMCID: PMC5460933 DOI: 10.1210/en.2016-1700] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 04/11/2017] [Indexed: 01/23/2023]
Abstract
The α-subunit of the heterotrimeric Gz protein, Gαz, promotes β-cell death and inhibits β-cell replication when pancreatic islets are challenged by stressors. Thus, we hypothesized that loss of Gαz protein would preserve functional β-cell mass in the nonobese diabetic (NOD) model, protecting from overt diabetes. We saw that protection from diabetes was robust and durable up to 35 weeks of age in Gαz knockout mice. By 17 weeks of age, Gαz-null NOD mice had significantly higher diabetes-free survival than wild-type littermates. Islets from these mice had reduced markers of proinflammatory immune cell infiltration on both the histological and transcript levels and secreted more insulin in response to glucose. Further analyses of pancreas sections revealed significantly fewer terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling (TUNEL)-positive β-cells in Gαz-null islets despite similar immune infiltration in control mice. Islets from Gαz-null mice also exhibited a higher percentage of Ki-67-positive β-cells, a measure of proliferation, even in the presence of immune infiltration. Finally, β-cell-specific Gαz-null mice phenocopy whole-body Gαz-null mice in their protection from developing hyperglycemia after streptozotocin administration, supporting a β-cell-centric role for Gαz in diabetes pathophysiology. We propose that Gαz plays a key role in β-cell signaling that becomes dysfunctional in the type 1 diabetes setting, accelerating the death of β-cells, which promotes further accumulation of immune cells in the pancreatic islets, and inhibiting a restorative proliferative response.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Blood Glucose/metabolism
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Female
- GTP-Binding Protein alpha Subunits/genetics
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, Transgenic
- Streptozocin
Collapse
Affiliation(s)
- Rachel J. Fenske
- Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Mark T. Cadena
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Quincy E. Harenda
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Haley N. Wienkes
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Kathryn Carbajal
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Michael D. Schaid
- Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Erin Laundre
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Allison L. Brill
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Nathan A. Truchan
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Harpreet Brar
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Jaclyn Wisinski
- Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Jinjin Cai
- Molecular Medicine Program, Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, Department of Nutrition, and Department of Biological Chemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah 84112
| | - Timothy E. Graham
- Molecular Medicine Program, Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, Department of Nutrition, and Department of Biological Chemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, Utah 84112
| | - Feyza Engin
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Michelle E. Kimple
- Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53705
| |
Collapse
|
22
|
Gregg T, Poudel C, Schmidt BA, Dhillon RS, Sdao SM, Truchan NA, Baar EL, Fernandez LA, Denu JM, Eliceiri KW, Rogers JD, Kimple ME, Lamming DW, Merrins MJ. Pancreatic β-Cells From Mice Offset Age-Associated Mitochondrial Deficiency With Reduced KATP Channel Activity. Diabetes 2016; 65:2700-10. [PMID: 27284112 PMCID: PMC5001174 DOI: 10.2337/db16-0432] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 05/31/2016] [Indexed: 12/20/2022]
Abstract
Aging is accompanied by impaired glucose homeostasis and an increased risk of type 2 diabetes, culminating in the failure of insulin secretion from pancreatic β-cells. To investigate the effects of age on β-cell metabolism, we established a novel assay to directly image islet metabolism with NAD(P)H fluorescence lifetime imaging (FLIM). We determined that impaired mitochondrial activity underlies an age-dependent loss of insulin secretion in human islets. NAD(P)H FLIM revealed a comparable decline in mitochondrial function in the pancreatic islets of aged mice (≥24 months), the result of 52% and 57% defects in flux through complex I and II, respectively, of the electron transport chain. However, insulin secretion and glucose tolerance are preserved in aged mouse islets by the heightened metabolic sensitivity of the β-cell triggering pathway, an adaptation clearly encoded in the metabolic and Ca(2+) oscillations that trigger insulin release (Ca(2+) plateau fraction: young 0.211 ± 0.006, aged 0.380 ± 0.007, P < 0.0001). This enhanced sensitivity is driven by a reduction in KATP channel conductance (diazoxide: young 5.1 ± 0.2 nS; aged 3.5 ± 0.5 nS, P < 0.01), resulting in an ∼2.8 mmol/L left shift in the β-cell glucose threshold. The results demonstrate how mice but not humans are able to successfully compensate for age-associated metabolic dysfunction by adjusting β-cell glucose sensitivity and highlight an essential mechanism for ensuring the maintenance of insulin secretion.
Collapse
Affiliation(s)
- Trillian Gregg
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI Biophysics Graduate Training Program, University of Wisconsin-Madison, Madison, WI Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI
| | - Chetan Poudel
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI
| | - Brian A Schmidt
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI
| | - Rashpal S Dhillon
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI
| | - Sophia M Sdao
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI
| | - Nathan A Truchan
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI
| | - Emma L Baar
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI
| | - Luis A Fernandez
- Department of Surgery, Division of Transplantation, University of Wisconsin-Madison, Madison, WI
| | - John M Denu
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI
| | - Kevin W Eliceiri
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI
| | - Jeremy D Rogers
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI
| | - Michelle E Kimple
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Dudley W Lamming
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI William S. Middleton Memorial Veterans Hospital, Madison, WI
| | - Matthew J Merrins
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin-Madison, Madison, WI Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, WI Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI William S. Middleton Memorial Veterans Hospital, Madison, WI
| |
Collapse
|
23
|
Fontana L, Cummings NE, Arriola Apelo SI, Neuman JC, Kasza I, Schmidt BA, Cava E, Spelta F, Tosti V, Syed FA, Baar EL, Veronese N, Cottrell SE, Fenske RJ, Bertozzi B, Brar HK, Pietka T, Bullock AD, Figenshau RS, Andriole GL, Merrins MJ, Alexander CM, Kimple ME, Lamming DW. Decreased Consumption of Branched-Chain Amino Acids Improves Metabolic Health. Cell Rep 2016; 16:520-530. [PMID: 27346343 DOI: 10.1016/j.celrep.2016.05.092] [Citation(s) in RCA: 336] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/27/2016] [Accepted: 05/26/2016] [Indexed: 11/28/2022] Open
Abstract
Protein-restricted (PR), high-carbohydrate diets improve metabolic health in rodents, yet the precise dietary components that are responsible for these effects have not been identified. Furthermore, the applicability of these studies to humans is unclear. Here, we demonstrate in a randomized controlled trial that a moderate PR diet also improves markers of metabolic health in humans. Intriguingly, we find that feeding mice a diet specifically reduced in branched-chain amino acids (BCAAs) is sufficient to improve glucose tolerance and body composition equivalently to a PR diet via metabolically distinct pathways. Our results highlight a critical role for dietary quality at the level of amino acids in the maintenance of metabolic health and suggest that diets specifically reduced in BCAAs, or pharmacological interventions in this pathway, may offer a translatable way to achieve many of the metabolic benefits of a PR diet.
Collapse
Affiliation(s)
- Luigi Fontana
- Division of Geriatrics and Nutritional Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Clinical and Experimental Sciences, University of Brescia Medical School, 25121 Brescia, Italy; CEINGE Biotecnologie Avanzate, 80145 Napoli, Italy.
| | - Nicole E Cummings
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sebastian I Arriola Apelo
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Joshua C Neuman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ildiko Kasza
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Brian A Schmidt
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Edda Cava
- Division of Geriatrics and Nutritional Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Francesco Spelta
- Division of Geriatrics and Nutritional Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Medicine, University of Verona, 37129 Verona, Italy
| | - Valeria Tosti
- Division of Geriatrics and Nutritional Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Medicine, University of Verona, 37129 Verona, Italy
| | - Faizan A Syed
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Emma L Baar
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Nicola Veronese
- Division of Geriatrics and Nutritional Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA; Division of Geriatrics, Department of Medicine, University of Padova, 35122 Padova, Italy
| | - Sara E Cottrell
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Rural and Urban Scholars in Community Health Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Rachel J Fenske
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Beatrice Bertozzi
- Division of Geriatrics and Nutritional Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Harpreet K Brar
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Terri Pietka
- Division of Geriatrics and Nutritional Sciences, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Arnold D Bullock
- Division of Urology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Robert S Figenshau
- Division of Urology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Gerald L Andriole
- Division of Urology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Matthew J Merrins
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Caroline M Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Michelle E Kimple
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA; Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53706, USA; Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; Rural and Urban Scholars in Community Health Program, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
24
|
Brill AL, Wisinski JA, Cadena MT, Thompson MF, Fenske RJ, Brar HK, Schaid MD, Pasker RL, Kimple ME. Synergy Between Gαz Deficiency and GLP-1 Analog Treatment in Preserving Functional β-Cell Mass in Experimental Diabetes. Mol Endocrinol 2016; 30:543-56. [PMID: 27049466 DOI: 10.1210/me.2015-1164] [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
A defining characteristic of type 1 diabetes mellitus (T1DM) pathophysiology is pancreatic β-cell death and dysfunction, resulting in insufficient insulin secretion to properly control blood glucose levels. Treatments that promote β-cell replication and survival, thus reversing the loss of β-cell mass, while also preserving β-cell function, could lead to a real cure for T1DM. The α-subunit of the heterotrimeric Gz protein, Gαz, is a tonic negative regulator of adenylate cyclase and downstream cAMP production. cAMP is one of a few identified signaling molecules that can simultaneously have a positive impact on pancreatic islet β-cell proliferation, survival, and function. The purpose of our study was to determine whether mice lacking Gαz might be protected, at least partially, from β-cell loss and dysfunction after streptozotocin treatment. We also aimed to determine whether Gαz might act in concert with an activator of the cAMP-stimulatory glucagon-like peptide 1 receptor, exendin-4 (Ex4). Without Ex4 treatment, Gαz-null mice still developed hyperglycemia, albeit delayed. The same finding held true for wild-type mice treated with Ex4. With Ex4 treatment, Gαz-null mice were protected from developing severe hyperglycemia. Immunohistological studies performed on pancreas sections and in vitro apoptosis, cytotoxicity, and survival assays demonstrated a clear effect of Gαz signaling on pancreatic β-cell replication and death; β-cell function was also improved in Gαz-null islets. These data support our hypothesis that a combination of therapies targeting both stimulatory and inhibitory pathways will be more effective than either alone at protecting, preserving, and possibly regenerating β-cell mass and function in T1DM.
Collapse
Affiliation(s)
- Allison L Brill
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Jaclyn A Wisinski
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Mark T Cadena
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Mary F Thompson
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Rachel J Fenske
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Harpreet K Brar
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Michael D Schaid
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Renee L Pasker
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Michelle E Kimple
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| |
Collapse
|
25
|
Lavine JA, Kibbe CR, Baan M, Sirinvaravong S, Umhoefer HM, Engler KA, Meske LM, Sacotte KA, Erhardt DP, Davis DB. Cholecystokinin expression in the β-cell leads to increased β-cell area in aged mice and protects from streptozotocin-induced diabetes and apoptosis. Am J Physiol Endocrinol Metab 2015; 309:E819-28. [PMID: 26394663 PMCID: PMC4652070 DOI: 10.1152/ajpendo.00159.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 09/15/2015] [Indexed: 12/23/2022]
Abstract
Cholecystokinin (CCK) is a peptide hormone produced in the gut and brain with beneficial effects on digestion, satiety, and insulin secretion. CCK is also expressed in pancreatic β-cells, but only in models of obesity and insulin resistance. Whole body deletion of CCK in obese mice leads to reduced β-cell mass expansion and increased apoptosis. We hypothesized that islet-derived CCK is important in protection from β-cell apoptosis. To determine the specific role of β-cell-derived CCK in β-cell mass dynamics, we generated a transgenic mouse that expresses CCK in the β-cell in the lean state (MIP-CCK). Although this transgene contains the human growth hormone minigene, we saw no expression of human growth hormone protein in transgenic islets. We examined the ability of MIP-CCK mice to maintain β-cell mass when subjected to apoptotic stress, with advanced age, and after streptozotocin treatment. Aged MIP-CCK mice have increased β-cell area. MIP-CCK mice are resistant to streptozotocin-induced diabetes and exhibit reduced β-cell apoptosis. Directed CCK overexpression in cultured β-cells also protects from cytokine-induced apoptosis. We have identified an important new paracrine/autocrine effect of CCK in protection of β-cells from apoptotic stress. Understanding the role of β-cell CCK adds to the emerging knowledge of classic gut peptides in intraislet signaling. CCK receptor agonists are being investigated as therapeutics for obesity and diabetes. While these agonists clearly have beneficial effects on body weight and insulin sensitivity in peripheral tissues, they may also directly protect β-cells from apoptosis.
Collapse
Affiliation(s)
- Jeremy A Lavine
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Carly R Kibbe
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Mieke Baan
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Sirinart Sirinvaravong
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Heidi M Umhoefer
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kimberly A Engler
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Louise M Meske
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kaitlyn A Sacotte
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Daniel P Erhardt
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Dawn Belt Davis
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin; Geriatric Research Education and Clinical Centers, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| |
Collapse
|