1
|
Kitamura RA, Hummel D, Ustione A, Piston DW, Urano F. Dual role of neuroplastin in pancreatic β cells: Regulating insulin secretion and promoting islet inflammation. Proc Natl Acad Sci U S A 2024; 121:e2411234121. [PMID: 39666939 PMCID: PMC11331099 DOI: 10.1073/pnas.2411234121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/03/2024] [Indexed: 12/14/2024] Open
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER)-resident secretory protein that reduces inflammation and promotes proliferation in pancreatic β cells. Numerous studies have highlighted the potential of MANF as a therapeutic agent for diabetes mellitus (DM), making it essential to understand the mechanisms underlying MANF's functions. In our previous search for a molecule that mediates MANF signaling, we identified Neuroplastin (NPTN) as a binding partner of MANF that localizes on the cell surface. However, the roles of NPTN in pancreatic β cells remain unclear. In this study, we generated β cell-specific Nptn knockout (KO) mice and conducted metabolic characterization. NPTN deficiency improved glucose tolerance by increasing insulin secretion and β cell mass in the pancreas. Moreover, proliferation and mitochondrial numbers in β cells increased in Nptn KO islets. These phenotypes resulted from elevated cytosolic Ca2+ levels and subsequent activation of downstream molecules. Simultaneously, we demonstrated that NPTN induces the expression of proinflammatory cytokines via the TRAF6-NF-κB axis in β cells. Additionally, NPTN deficiency conferred resistance to streptozotocin-induced diabetic phenotypes. Finally, exogenous MANF treatment in islets or β cells led to similar phenotypes as those observed in NPTN-deficient models. These results indicate that NPTN plays important roles in the regulation of insulin secretion, proliferation, and mitochondrial quantity, as well as proinflammatory responses, which are antagonized by MANF treatment. Thus, targeting the MANF-NPTN interaction may lead to a novel treatment for improving β cell functions in DM.
Collapse
Affiliation(s)
- Rie Asada Kitamura
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO63110
| | - Devynn Hummel
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO63110
| | - Alessandro Ustione
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO63110
| | - David W. Piston
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO63110
| | - Fumihiko Urano
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO63110
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO63110
| |
Collapse
|
2
|
Jones Lipinski RA, Stancill JS, Nuñez R, Wynia-Smith SL, Sprague DJ, Nord JA, Bird A, Corbett JA, Smith BC. Zinc-chelating BET bromodomain inhibitors equally target islet endocrine cell types. Am J Physiol Regul Integr Comp Physiol 2024; 326:R515-R527. [PMID: 38618911 PMCID: PMC11381023 DOI: 10.1152/ajpregu.00259.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 03/19/2024] [Accepted: 04/07/2024] [Indexed: 04/16/2024]
Abstract
Inhibition of the bromodomain and extraterminal domain (BET) protein family is a potential strategy to prevent and treat diabetes; however, the clinical use of BET bromodomain inhibitors (BETis) is associated with adverse effects. Here, we explore a strategy for targeting BETis to β cells by exploiting the high-zinc (Zn2+) concentration in β cells relative to other cell types. We report the synthesis of a novel, Zn2+-chelating derivative of the pan-BETi (+)-JQ1, (+)-JQ1-DPA, in which (+)-JQ1 was conjugated to dipicolyl amine (DPA). As controls, we synthesized (+)-JQ1-DBA, a non-Zn2+-chelating derivative, and (-)-JQ1-DPA, an inactive enantiomer that chelates Zn2+. Molecular modeling and biophysical assays showed that (+)-JQ1-DPA and (+)-JQ1-DBA retain potent binding to BET bromodomains in vitro. Cellular assays demonstrated (+)-JQ1-DPA attenuated NF-ĸB target gene expression in β cells stimulated with the proinflammatory cytokine interleukin 1β. To assess β-cell selectivity, we isolated islets from a mouse model that expresses green fluorescent protein in insulin-positive β cells and mTomato in insulin-negative cells (non-β cells). Surprisingly, Zn2+ chelation did not confer β-cell selectivity as (+)-JQ1-DPA was equally effective in both β and α cells; however, (+)-JQ1-DPA was less effective in macrophages, a nonendocrine islet cell type. Intriguingly, the non-Zn2+-chelating derivative (+)-JQ1-DBA displayed the opposite selectivity, with greater effect in macrophages compared with (+)-JQ1-DPA, suggesting potential as a macrophage-targeting molecule. These findings suggest that Zn2+-chelating small molecules confer endocrine cell selectivity rather than β-cell selectivity in pancreatic islets and provide valuable insights and techniques to assess Zn2+ chelation as an approach to selectively target small molecules to pancreatic β cells.NEW & NOTEWORTHY Inhibition of BET bromodomains is a novel potential strategy to prevent and treat diabetes mellitus. However, BET inhibitors have negative side effects. We synthesized a BET inhibitor expected to exploit the high zinc concentration in β cells to accumulate in β cells. We show our inhibitor targeted pancreatic endocrine cells; however, it was less effective in immune cells. A control inhibitor showed the opposite effect. These findings help us understand how to target specific cells in diabetes treatment.
Collapse
Affiliation(s)
- Rachel A Jones Lipinski
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Jennifer S Stancill
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Raymundo Nuñez
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Sarah L Wynia-Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Daniel J Sprague
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Joshua A Nord
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Amir Bird
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Brian C Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
3
|
Zhang Q, Yang Z, Yang J. Dissecting the colocalized GWAS and eQTLs with mediation analysis for high-dimensional exposures and confounders. Biometrics 2024; 80:ujae050. [PMID: 38801257 DOI: 10.1093/biomtc/ujae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/14/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024]
Abstract
To leverage the advancements in genome-wide association studies (GWAS) and quantitative trait loci (QTL) mapping for traits and molecular phenotypes to gain mechanistic understanding of the genetic regulation, biological researchers often investigate the expression QTLs (eQTLs) that colocalize with QTL or GWAS peaks. Our research is inspired by 2 such studies. One aims to identify the causal single nucleotide polymorphisms that are responsible for the phenotypic variation and whose effects can be explained by their impacts at the transcriptomic level in maize. The other study in mouse focuses on uncovering the cis-driver genes that induce phenotypic changes by regulating trans-regulated genes. Both studies can be formulated as mediation problems with potentially high-dimensional exposures, confounders, and mediators that seek to estimate the overall indirect effect (IE) for each exposure. In this paper, we propose MedDiC, a novel procedure to estimate the overall IE based on difference-in-coefficients approach. Our simulation studies find that MedDiC offers valid inference for the IE with higher power, shorter confidence intervals, and faster computing time than competing methods. We apply MedDiC to the 2 aforementioned motivating datasets and find that MedDiC yields reproducible outputs across the analysis of closely related traits, with results supported by external biological evidence. The code and additional information are available on our GitHub page (https://github.com/QiZhangStat/MedDiC).
Collapse
Affiliation(s)
- Qi Zhang
- Department of Mathematics and Statistics, University of New Hampshire, Durham, NH 03824, United States
| | - Zhikai Yang
- Complex Biosystems Program and Department of Agronomy and Horticulture, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Jinliang Yang
- Department of Agronomy and Horticulture, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| |
Collapse
|
4
|
Keller MP, Hudkins KL, Shalev A, Bhatnagar S, Kebede MA, Merrins MJ, Davis DB, Alpers CE, Kimple ME, Attie AD. What the BTBR/J mouse has taught us about diabetes and diabetic complications. iScience 2023; 26:107036. [PMID: 37360692 PMCID: PMC10285641 DOI: 10.1016/j.isci.2023.107036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
Human and mouse genetics have delivered numerous diabetogenic loci, but it is mainly through the use of animal models that the pathophysiological basis for their contribution to diabetes has been investigated. More than 20 years ago, we serendipidously identified a mouse strain that could serve as a model of obesity-prone type 2 diabetes, the BTBR (Black and Tan Brachyury) mouse (BTBR T+ Itpr3tf/J, 2018) carrying the Lepob mutation. We went on to discover that the BTBR-Lepob mouse is an excellent model of diabetic nephropathy and is now widely used by nephrologists in academia and the pharmaceutical industry. In this review, we describe the motivation for developing this animal model, the many genes identified and the insights about diabetes and diabetes complications derived from >100 studies conducted in this remarkable animal model.
Collapse
Affiliation(s)
- Mark P. Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kelly L. Hudkins
- Department of Pathology, University of Washington Medical Center, Seattle, WA 98195, USA
| | - Anath Shalev
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, UK
| | - Sushant Bhatnagar
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294, UK
| | - Melkam A. Kebede
- School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Camperdown, Sydney, NSW 2006, Australia
| | - Matthew J. Merrins
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Dawn Belt Davis
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Charles E. Alpers
- Department of Pathology, University of Washington Medical Center, Seattle, WA 98195, USA
| | - Michelle E. Kimple
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Alan D. Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
5
|
Chella Krishnan K, El Hachem EJ, Keller MP, Patel SG, Carroll L, Vegas AD, Gerdes Gyuricza I, Light C, Cao Y, Pan C, Kaczor-Urbanowicz KE, Shravah V, Anum D, Pellegrini M, Lee CF, Seldin MM, Rosenthal NA, Churchill GA, Attie AD, Parker B, James DE, Lusis AJ. Genetic architecture of heart mitochondrial proteome influencing cardiac hypertrophy. eLife 2023; 12:e82619. [PMID: 37276142 PMCID: PMC10241513 DOI: 10.7554/elife.82619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 05/18/2023] [Indexed: 06/07/2023] Open
Abstract
Mitochondria play an important role in both normal heart function and disease etiology. We report analysis of common genetic variations contributing to mitochondrial and heart functions using an integrative proteomics approach in a panel of inbred mouse strains called the Hybrid Mouse Diversity Panel (HMDP). We performed a whole heart proteome study in the HMDP (72 strains, n=2-3 mice) and retrieved 848 mitochondrial proteins (quantified in ≥50 strains). High-resolution association mapping on their relative abundance levels revealed three trans-acting genetic loci on chromosomes (chr) 7, 13 and 17 that regulate distinct classes of mitochondrial proteins as well as cardiac hypertrophy. DAVID enrichment analyses of genes regulated by each of the loci revealed that the chr13 locus was highly enriched for complex-I proteins (24 proteins, P=2.2E-61), the chr17 locus for mitochondrial ribonucleoprotein complex (17 proteins, P=3.1E-25) and the chr7 locus for ubiquinone biosynthesis (3 proteins, P=6.9E-05). Follow-up high resolution regional mapping identified NDUFS4, LRPPRC and COQ7 as the candidate genes for chr13, chr17 and chr7 loci, respectively, and both experimental and statistical analyses supported their causal roles. Furthermore, a large cohort of Diversity Outbred mice was used to corroborate Lrpprc gene as a driver of mitochondrial DNA (mtDNA)-encoded gene regulation, and to show that the chr17 locus is specific to heart. Variations in all three loci were associated with heart mass in at least one of two independent heart stress models, namely, isoproterenol-induced heart failure and diet-induced obesity. These findings suggest that common variations in certain mitochondrial proteins can act in trans to influence tissue-specific mitochondrial functions and contribute to heart hypertrophy, elucidating mechanisms that may underlie genetic susceptibility to heart failure in human populations.
Collapse
Affiliation(s)
- Karthickeyan Chella Krishnan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of MedicineCincinnatiUnited States
| | - Elie-Julien El Hachem
- Department of Integrative Biology and Physiology, Field Systems Biology, Sciences Sorbonne UniversitéParisFrance
| | - Mark P Keller
- Biochemistry Department, University of Wisconsin-MadisonMadisonUnited States
| | - Sanjeet G Patel
- Department of Surgery/Division of Cardiac Surgery, University of Southern California Keck School of MedicineLos AngelesUnited States
| | - Luke Carroll
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Alexis Diaz Vegas
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | | | - Christine Light
- Cardiovascular Biology Research Program, Oklahoma Medical Research FoundationOklahoma CityUnited States
| | - Yang Cao
- Department of Medicine/Division of Cardiology, University of California, Los AngelesLos AngelesUnited States
| | - Calvin Pan
- Department of Medicine/Division of Cardiology, University of California, Los AngelesLos AngelesUnited States
| | - Karolina Elżbieta Kaczor-Urbanowicz
- Division of Oral Biology and Medicine, UCLA School of DentistryLos AngelesUnited States
- UCLA Institute for Quantitative and Computational BiosciencesLos AngelesUnited States
| | - Varun Shravah
- Department of Chemistry, University of CaliforniaLos AngelesUnited States
| | - Diana Anum
- Department of Integrative Biology and Physiology, University of CaliforniaLos AngelesUnited States
| | - Matteo Pellegrini
- UCLA Institute for Quantitative and Computational BiosciencesLos AngelesUnited States
| | - Chi Fung Lee
- Cardiovascular Biology Research Program, Oklahoma Medical Research FoundationOklahoma CityUnited States
- Department of Physiology, University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Marcus M Seldin
- Center for Epigenetics and MetabolismIrvineUnited States
- Department of Biological Chemistry, University of CaliforniaIrvineUnited States
| | | | | | - Alan D Attie
- Biochemistry Department, University of Wisconsin-MadisonMadisonUnited States
| | - Benjamin Parker
- Department of Anatomy and Physiology, University of MelbourneMelbourneAustralia
| | - David E James
- Metabolic Systems Biology Laboratory, Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneySydneyAustralia
| | - Aldons J Lusis
- Department of Medicine/Division of Cardiology, University of California, Los AngelesLos AngelesUnited States
- Department of Human Genetics, University of CaliforniaLos AngelesUnited States
- Department of Microbiology, Immunology and Molecular Genetics, University of CaliforniaLos AngelesUnited States
| |
Collapse
|
6
|
Benaglio P, Zhu H, Okino ML, Yan J, Elgamal R, Nariai N, Beebe E, Korgaonkar K, Qiu Y, Donovan MK, Chiou J, Wang G, Newsome J, Kaur J, Miller M, Preissl S, Corban S, Aylward A, Taipale J, Ren B, Frazer KA, Sander M, Gaulton KJ. Type 1 diabetes risk genes mediate pancreatic beta cell survival in response to proinflammatory cytokines. CELL GENOMICS 2022; 2:100214. [PMID: 36778047 PMCID: PMC9903835 DOI: 10.1016/j.xgen.2022.100214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 06/17/2022] [Accepted: 10/15/2022] [Indexed: 11/13/2022]
Abstract
We combined functional genomics and human genetics to investigate processes that affect type 1 diabetes (T1D) risk by mediating beta cell survival in response to proinflammatory cytokines. We mapped 38,931 cytokine-responsive candidate cis-regulatory elements (cCREs) in beta cells using ATAC-seq and snATAC-seq and linked them to target genes using co-accessibility and HiChIP. Using a genome-wide CRISPR screen in EndoC-βH1 cells, we identified 867 genes affecting cytokine-induced survival, and genes promoting survival and up-regulated in cytokines were enriched at T1D risk loci. Using SNP-SELEX, we identified 2,229 variants in cytokine-responsive cCREs altering transcription factor (TF) binding, and variants altering binding of TFs regulating stress, inflammation, and apoptosis were enriched for T1D risk. At the 16p13 locus, a fine-mapped T1D variant altering TF binding in a cytokine-induced cCRE interacted with SOCS1, which promoted survival in cytokine exposure. Our findings reveal processes and genes acting in beta cells during inflammation that modulate T1D risk.
Collapse
Affiliation(s)
- Paola Benaglio
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Han Zhu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Mei-Lin Okino
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jian Yan
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
- School of Medicine, Northwest University, Xi’an, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
| | - Ruth Elgamal
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Naoki Nariai
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Elisha Beebe
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Katha Korgaonkar
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Yunjiang Qiu
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | | | - Joshua Chiou
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Gaowei Wang
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jacklyn Newsome
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Jaspreet Kaur
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Michael Miller
- Center for Epigenomics, University of California, San Diego, La Jolla, CA, USA
| | - Sebastian Preissl
- Center for Epigenomics, University of California, San Diego, La Jolla, CA, USA
| | - Sierra Corban
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Anthony Aylward
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Jussi Taipale
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Genome-Scale Biology Program, University of Helsinki, Helsinki, Finland
| | - Bing Ren
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
- Center for Epigenomics, University of California, San Diego, La Jolla, CA, USA
| | - Kelly A. Frazer
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Maike Sander
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Kyle J. Gaulton
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
7
|
Wakeling MN, Owens NDL, Hopkinson JR, Johnson MB, Houghton JAL, Dastamani A, Flaxman CS, Wyatt RC, Hewat TI, Hopkins JJ, Laver TW, van Heugten R, Weedon MN, De Franco E, Patel KA, Ellard S, Morgan NG, Cheesman E, Banerjee I, Hattersley AT, Dunne MJ, Richardson SJ, Flanagan SE. Non-coding variants disrupting a tissue-specific regulatory element in HK1 cause congenital hyperinsulinism. Nat Genet 2022; 54:1615-1620. [PMID: 36333503 PMCID: PMC7614032 DOI: 10.1038/s41588-022-01204-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 09/16/2022] [Indexed: 11/06/2022]
Abstract
Gene expression is tightly regulated, with many genes exhibiting cell-specific silencing when their protein product would disrupt normal cellular function1. This silencing is largely controlled by non-coding elements, and their disruption might cause human disease2. We performed gene-agnostic screening of the non-coding regions to discover new molecular causes of congenital hyperinsulinism. This identified 14 non-coding de novo variants affecting a 42-bp conserved region encompassed by a regulatory element in intron 2 of the hexokinase 1 gene (HK1). HK1 is widely expressed across all tissues except in the liver and pancreatic beta cells and is thus termed a 'disallowed gene' in these specific tissues. We demonstrated that the variants result in a loss of repression of HK1 in pancreatic beta cells, thereby causing insulin secretion and congenital hyperinsulinism. Using epigenomic data accessed from public repositories, we demonstrated that these variants reside within a regulatory region that we determine to be critical for cell-specific silencing. Importantly, this has revealed a disease mechanism for non-coding variants that cause inappropriate expression of a disallowed gene.
Collapse
Affiliation(s)
- Matthew N Wakeling
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Nick D L Owens
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Jessica R Hopkinson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Matthew B Johnson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Jayne A L Houghton
- Exeter Genomics Laboratory, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Antonia Dastamani
- Endocrinology Department, Great Ormond Street Hospital for Children, London, UK
| | - Christine S Flaxman
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Rebecca C Wyatt
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Thomas I Hewat
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Jasmin J Hopkins
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Thomas W Laver
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Rachel van Heugten
- Exeter Genomics Laboratory, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Michael N Weedon
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Elisa De Franco
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Kashyap A Patel
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Sian Ellard
- Exeter Genomics Laboratory, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Noel G Morgan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Edmund Cheesman
- Department of Paediatric Pathology, Royal Manchester Children's Hospital, Manchester, UK
| | - Indraneel Banerjee
- Department of Paediatric Endocrinology, Royal Manchester Children's Hospital, Manchester, UK
- Faculty of Biology, Medicine and Health, the University of Manchester, Manchester, UK
| | - Andrew T Hattersley
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Mark J Dunne
- Faculty of Biology, Medicine and Health, the University of Manchester, Manchester, UK
| | - Sarah J Richardson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Sarah E Flanagan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
8
|
Alghamdi TA, Krentz NA, Smith N, Spigelman AF, Rajesh V, Jha A, Ferdaoussi M, Suzuki K, Yang J, Manning Fox JE, Sun H, Sun Z, Gloyn AL, MacDonald PE. Zmiz1 is required for mature β-cell function and mass expansion upon high fat feeding. Mol Metab 2022; 66:101621. [PMID: 36307047 PMCID: PMC9643564 DOI: 10.1016/j.molmet.2022.101621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/15/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Identifying the transcripts which mediate genetic association signals for type 2 diabetes (T2D) is critical to understand disease mechanisms. Studies in pancreatic islets support the transcription factor ZMIZ1 as a transcript underlying a T2D GWAS signal, but how it influences T2D risk is unknown. METHODS β-Cell-specific Zmiz1 knockout (Zmiz1βKO) mice were generated and phenotypically characterised. Glucose homeostasis was assessed in Zmiz1βKO mice and their control littermates on chow diet (CD) and high fat diet (HFD). Islet morphology and function were examined by immunohistochemistry and in vitro islet function was assessed by dynamic insulin secretion assay. Transcript and protein expression were assessed by RNA sequencing and Western blotting. In islets isolated from genotyped human donors, we assessed glucose-dependent insulin secretion and islet insulin content by static incubation assay. RESULTS Male and female Zmiz1βKO mice were glucose intolerant with impaired insulin secretion, compared with control littermates. Transcriptomic profiling of Zmiz1βKO islets identified over 500 differentially expressed genes including those involved in β-cell function and maturity, which we confirmed at the protein level. Upon HFD, Zmiz1βKO mice fail to expand β-cell mass and become severely diabetic. Human islets from carriers of the ZMIZ1-linked T2D-risk alleles have reduced islet insulin content and glucose-stimulated insulin secretion. CONCLUSIONS β-Cell Zmiz1 is required for normal glucose homeostasis. Genetic variation at the ZMIZ1 locus may influence T2D-risk by reducing islet mass expansion upon metabolic stress and the ability to maintain a mature β-cell state.
Collapse
Affiliation(s)
- Tamadher A. Alghamdi
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, T6G2R3, Canada
| | - Nicole A.J. Krentz
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Nancy Smith
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, T6G2R3, Canada
| | - Aliya F. Spigelman
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, T6G2R3, Canada
| | - Varsha Rajesh
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alokkumar Jha
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Mourad Ferdaoussi
- Department of Pediatrics, University of Alberta, Edmonton AB, T6G2R3, Canada
| | - Kunimasa Suzuki
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, T6G2R3, Canada
| | - Jing Yang
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jocelyn E. Manning Fox
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, T6G2R3, Canada
| | - Han Sun
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Zijie Sun
- Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Anna L. Gloyn
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA,Stanford Diabetes Research Centre, Stanford University, Stanford, CA, USA,Oxford Centre for Diabetes Endocrinology & Metabolism, Radcliffe Department of Medicine, University of Oxford, UK,Corresponding author. Center for Academic Medicine, Division of Endocrinology & Diabetes, Department of Pediatrics, 453 Quarry Road, Palo Alto CA, 94304, USA. http://www.bcell.org
| | - Patrick E. MacDonald
- Department of Pharmacology and Alberta Diabetes Institute, University of Alberta, Edmonton, AB, T6G2R3, Canada,Corresponding author. Alberta Diabetes Institute, LKS Centre, Rm. 6-126, Edmonton, AB, T6G 2R3, Canada.
| |
Collapse
|
9
|
Wang P, Karakose E, Argmann C, Wang H, Balev M, Brody RI, Rivas HG, Liu X, Wood O, Liu H, Choleva L, Hasson D, Bernstein E, Paulo JA, Scott DK, Lambertini L, DeCaprio JA, Stewart AF. Disrupting the DREAM complex enables proliferation of adult human pancreatic β cells. J Clin Invest 2022; 132:e157086. [PMID: 35700053 PMCID: PMC9337832 DOI: 10.1172/jci157086] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/09/2022] [Indexed: 11/17/2022] Open
Abstract
Resistance to regeneration of insulin-producing pancreatic β cells is a fundamental challenge for type 1 and type 2 diabetes. Recently, small molecule inhibitors of the kinase DYRK1A have proven effective in inducing adult human β cells to proliferate, but their detailed mechanism of action is incompletely understood. We interrogated our human insulinoma and β cell transcriptomic databases seeking to understand why β cells in insulinomas proliferate, while normal β cells do not. This search reveals the DREAM complex as a central regulator of quiescence in human β cells. The DREAM complex consists of a module of transcriptionally repressive proteins that assemble in response to DYRK1A kinase activity, thereby inducing and maintaining cellular quiescence. In the absence of DYRK1A, DREAM subunits reassemble into the pro-proliferative MMB complex. Here, we demonstrate that small molecule DYRK1A inhibitors induce human β cells to replicate by converting the repressive DREAM complex to its pro-proliferative MMB conformation.
Collapse
Affiliation(s)
- Peng Wang
- Diabetes Obesity Metabolism Institute
- Department of Medicine, and
| | - Esra Karakose
- Diabetes Obesity Metabolism Institute
- Department of Medicine, and
| | - Carmen Argmann
- Department of Genetics and Genomic Sciences, The Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | - Rachel I. Brody
- Department of Pathology, The Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hembly G. Rivas
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- The Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xinyue Liu
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Olivia Wood
- Diabetes Obesity Metabolism Institute
- Department of Medicine, and
| | - Hongtao Liu
- Diabetes Obesity Metabolism Institute
- Department of Medicine, and
| | - Lauryn Choleva
- Diabetes Obesity Metabolism Institute
- Department of Pediatrics
| | - Dan Hasson
- The Tisch Cancer Institute
- Department of Oncological Sciences
- Bioinformatics for Next Generation Sequencing (BiNGS) Shared Resource Facility, and
| | - Emily Bernstein
- The Tisch Cancer Institute
- Department of Oncological Sciences
- The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Joao A. Paulo
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Donald K. Scott
- Diabetes Obesity Metabolism Institute
- Department of Medicine, and
| | - Luca Lambertini
- Diabetes Obesity Metabolism Institute
- Department of Medicine, and
| | - James A. DeCaprio
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- The Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
10
|
Xie Y, Cui Z, Wang N, Li P. Research on Potential Network Markers and Signaling Pathways in Type 2 Diabetes Based on Conditional Cell-Specific Network. Genes (Basel) 2022; 13:1155. [PMID: 35885938 PMCID: PMC9320152 DOI: 10.3390/genes13071155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022] Open
Abstract
Traditional methods concerning type 2 diabetes (T2D) are limited to grouped cells instead of each single cell, and thus the heterogeneity of single cells is erased. Therefore, it is still challenging to study T2D based on a single-cell and network perspective. In this study, we construct a conditional cell-specific network (CCSN) for each single cell for the GSE86469 dataset which is a single-cell transcriptional set from nondiabetic (ND) and T2D human islet samples, and obtain a conditional network degree matrix (CNDM). Since beta cells are the key cells leading to T2D, we search for hub genes in CCSN of beta cells and find that ATP6AP2 is essential for regulation and storage of insulin, and the renin-angiotensin system involving ATP6AP2 is related to most pathological processes leading to diabetic nephropathy. The communication between beta cells and other endocrine cells is performed and three gene pairs with obvious interaction are found. In addition, different expression genes (DEGs) are found based on CNDM and the gene expression matrix (GEM), respectively. Finally, 'dark' genes are identified, and enrichment analysis shows that NFATC2 is involved in the VEGF signaling pathway and indirectly affects the production of Prostacyclin (PGI2), which may be a potential biomarker for diabetic nephropathy.
Collapse
Affiliation(s)
| | | | | | - Peiluan Li
- School of Mathematics and Statistics, Henan University of Science and Technology, Luoyang 471023, China; (Y.X.); (Z.C.); (N.W.)
| |
Collapse
|
11
|
Abderrahmani A, Jacovetti C, Regazzi R. Lessons from neonatal β-cell epigenomic for diabetes prevention and treatment. Trends Endocrinol Metab 2022; 33:378-389. [PMID: 35382967 DOI: 10.1016/j.tem.2022.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 12/30/2022]
Abstract
Pancreatic β-cell expansion and functional maturation during the birth-to-weaning period plays an essential role in the adaptation of plasma insulin levels to metabolic needs. These events are driven by epigenetic programs triggered by growth factors, hormones, and nutrients. These mechanisms operating in the neonatal period can be at least in part reactivated in adult life to increase the functional β-cell mass and face conditions of increased insulin demand such as obesity or pregnancy. In this review, we will highlight the importance of studying these signaling pathways and epigenetic programs to understand the causes of different forms of diabetes and to permit the design of novel therapeutic strategies to prevent and treat this metabolic disorder affecting hundreds of millions of people worldwide.
Collapse
Affiliation(s)
- Amar Abderrahmani
- Universitéde Lille, CNRS, Centrale Lille, Université Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| | - Cécile Jacovetti
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Romano Regazzi
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland; Department of Biomedical Science, University of Lausanne, 1005 Lausanne, Switzerland.
| |
Collapse
|
12
|
Darden CM, Vasu S, Mattke J, Liu Y, Rhodes CJ, Naziruddin B, Lawrence MC. Calcineurin/NFATc2 and PI3K/AKT signaling maintains β-cell identity and function during metabolic and inflammatory stress. iScience 2022; 25:104125. [PMID: 35402865 PMCID: PMC8983383 DOI: 10.1016/j.isci.2022.104125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/02/2021] [Accepted: 03/16/2022] [Indexed: 11/19/2022] Open
Abstract
Pancreatic islets respond to metabolic and inflammatory stress by producing hormones and other factors that induce adaptive cellular and systemic responses. Here we show that intracellular Ca2+ ([Ca2+]i) and ROS signals generated by high glucose and cytokine-induced ER stress activate calcineurin (CN)/NFATc2 and PI3K/AKT to maintain β-cell identity and function. This was attributed in part by direct induction of the endocrine differentiation gene RFX6 and suppression of several β-cell "disallowed" genes, including MCT1. CN/NFATc2 targeted p300 and HDAC1 to RFX6 and MCT1 promoters to induce and suppress gene transcription, respectively. In contrast, prolonged exposure to stress, hyperstimulated [Ca2+]i, or perturbation of CN/NFATc2 resulted in downregulation of RFX6 and induction of MCT1. These findings reveal that CN/NFATc2 and PI3K/AKT maintain β-cell function during acute stress, but β-cells dedifferentiate to a dysfunctional state upon loss or exhaustion of Ca2+/CN/NFATc2 signaling. They further demonstrate the utility of targeting CN/NFATc2 to restore β-cell function.
Collapse
Affiliation(s)
- Carly M. Darden
- Islet Cell Laboratory, Baylor Scott & White Research Institute, Dallas, TX 75204, USA
- Institute of Biomedical Studies, Baylor University, Waco, TX 76706, USA
| | - Srividya Vasu
- Islet Cell Laboratory, Baylor Scott & White Research Institute, Dallas, TX 75204, USA
| | - Jordan Mattke
- Islet Cell Laboratory, Baylor Scott & White Research Institute, Dallas, TX 75204, USA
- Institute of Biomedical Studies, Baylor University, Waco, TX 76706, USA
| | - Yang Liu
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX 75246, USA
| | - Christopher J. Rhodes
- Kovler Diabetes Center, Department of Medicine, Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, IL 60637, USA
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca Ltd, Gaithersburg, MD 20878, USA
| | - Bashoo Naziruddin
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX 75246, USA
| | - Michael C. Lawrence
- Islet Cell Laboratory, Baylor Scott & White Research Institute, Dallas, TX 75204, USA
| |
Collapse
|
13
|
Miranda JG, Schleicher WE, Wells KL, Ramirez DG, Landgrave SP, Benninger RKP. Dynamic changes in β-cell [Ca 2+] regulate NFAT activation, gene transcription, and islet gap junction communication. Mol Metab 2022; 57:101430. [PMID: 34979329 PMCID: PMC8804269 DOI: 10.1016/j.molmet.2021.101430] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Diabetes occurs because of insufficient insulin secretion due to β-cell dysfunction within the islet of Langerhans. Elevated glucose levels trigger β-cell membrane depolarization, action potential generation, and slow sustained free-Ca2+ ([Ca2+]) oscillations, which trigger insulin release. Nuclear factor of activated T-cell (NFAT) is a transcription factor, which is regulated by the increases in [Ca2+] and calceineurin (CaN) activation. NFAT regulation links cell activity with gene transcription in many systems and regulates proliferation and insulin granule biogenesis within the β-cell. However, the link between the regulation of β-cell electrical activity and oscillatory [Ca2+] dynamics with NFAT activation and downstream transcription is poorly understood. Here, we tested whether dynamic changes to β-cell electrical activity and [Ca2+] regulate NFAT activation and downstream transcription. METHODS In cell lines, mouse islets, and human islets, including those from donors with type 2 diabetes, we applied both agonists/antagonists of ion channels together with optogenetics to modulate β-cell electrical activity. We measured the dynamics of [Ca2+] and NFAT activation as well as performed whole transcriptome and functional analyses. RESULTS Both glucose-induced membrane depolarization and optogenetic stimulation triggered NFAT activation as well as increased the transcription of NFAT targets and intermediate early genes (IEGs). Importantly, slow, sustained [Ca2+] oscillation conditions led to NFAT activation and downstream transcription. In contrast, in human islets from donors with type2 diabetes, NFAT activation by glucose was diminished, but rescued upon pharmacological stimulation of electrical activity. NFAT activation regulated GJD2 expression and increased Cx36 gap junction permeability upon elevated oscillatory [Ca2+] dynamics. However, it is unclear if NFAT directly binds the GJD2 gene to regulate expression. CONCLUSIONS This study provides an insight into the specific patterns of electrical activity that regulate NFAT activation, gene transcription, and islet function. In addition, it provides information on how these factors are disrupted in diabetes.
Collapse
Affiliation(s)
- Jose G Miranda
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora CO, 80045, USA
| | - Wolfgang E Schleicher
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora CO, 80045, USA
| | - Kristen L Wells
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - David G Ramirez
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora CO, 80045, USA
| | - Samantha P Landgrave
- Program in Cell Biology, Stem Cell and Development, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Richard K P Benninger
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora CO, 80045, USA; Program in Cell Biology, Stem Cell and Development, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA; Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
14
|
Joshi H, Vastrad B, Joshi N, Vastrad C. Integrated bioinformatics analysis reveals novel key biomarkers in diabetic nephropathy. SAGE Open Med 2022; 10:20503121221137005. [PMID: 36385790 PMCID: PMC9661593 DOI: 10.1177/20503121221137005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives: The underlying molecular mechanisms of diabetic nephropathy have yet not been investigated clearly. In this investigation, we aimed to identify key genes involved in the pathogenesis and prognosis of diabetic nephropathy. Methods: We downloaded next-generation sequencing data set GSE142025 from Gene Expression Omnibus database having 28 diabetic nephropathy samples and nine normal control samples. The differentially expressed genes between diabetic nephropathy and normal control samples were analyzed. Biological function analysis of the differentially expressed genes was enriched by Gene Ontology and REACTOME pathways. Then, we established the protein–protein interaction network, modules, miRNA-differentially expressed gene regulatory network and transcription factor-differentially expressed gene regulatory network. Hub genes were validated by using receiver operating characteristic curve analysis. Results: A total of 549 differentially expressed genes were detected including 275 upregulated and 274 downregulated genes. The biological process analysis of functional enrichment showed that these differentially expressed genes were mainly enriched in cell activation, integral component of plasma membrane, lipid binding, and biological oxidations. Analyzing the protein–protein interaction network, miRNA-differentially expressed gene regulatory network and transcription factor-differentially expressed gene regulatory network, we screened hub genes MDFI, LCK, BTK, IRF4, PRKCB, EGR1, JUN, FOS, ALB, and NR4A1 by the Cytoscape software. The receiver operating characteristic curve analysis confirmed that hub genes were of diagnostic value. Conclusions: Taken above, using integrated bioinformatics analysis, we have identified key genes and pathways in diabetic nephropathy, which could improve our understanding of the cause and underlying molecular events, and these key genes and pathways might be therapeutic targets for diabetic nephropathy.
Collapse
Affiliation(s)
- Harish Joshi
- Endocrine and Diabetes Care Center, Hubbali, India
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry, KLE Society’s College of Pharmacy, Gadag, India
| | - Nidhi Joshi
- Dr. D. Y. Patil Medical College, Kolhapur, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Dharwad, India
- Chanabasayya Vastrad, Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, India.
| |
Collapse
|
15
|
Simonett SP, Shin S, Herring JA, Bacher R, Smith LA, Dong C, Rabaglia ME, Stapleton DS, Schueler KL, Choi J, Bernstein MN, Turkewitz DR, Perez-Cervantes C, Spaeth J, Stein R, Tessem JS, Kendziorski C, Keleş S, Moskowitz IP, Keller MP, Attie AD. Identification of direct transcriptional targets of NFATC2 that promote β cell proliferation. J Clin Invest 2021; 131:e144833. [PMID: 34491912 PMCID: PMC8553569 DOI: 10.1172/jci144833] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 09/02/2021] [Indexed: 12/13/2022] Open
Abstract
The transcription factor NFATC2 induces β cell proliferation in mouse and human islets. However, the genomic targets that mediate these effects have not been identified. We expressed active forms of Nfatc2 and Nfatc1 in human islets. By integrating changes in gene expression with genomic binding sites for NFATC2, we identified approximately 2200 transcriptional targets of NFATC2. Genes induced by NFATC2 were enriched for transcripts that regulate the cell cycle and for DNA motifs associated with the transcription factor FOXP. Islets from an endocrine-specific Foxp1, Foxp2, and Foxp4 triple-knockout mouse were less responsive to NFATC2-induced β cell proliferation, suggesting the FOXP family works to regulate β cell proliferation in concert with NFATC2. NFATC2 induced β cell proliferation in both mouse and human islets, whereas NFATC1 did so only in human islets. Exploiting this species difference, we identified approximately 250 direct transcriptional targets of NFAT in human islets. This gene set enriches for cell cycle-associated transcripts and includes Nr4a1. Deletion of Nr4a1 reduced the capacity of NFATC2 to induce β cell proliferation, suggesting that much of the effect of NFATC2 occurs through its induction of Nr4a1. Integration of noncoding RNA expression, chromatin accessibility, and NFATC2 binding sites enabled us to identify NFATC2-dependent enhancer loci that mediate β cell proliferation.
Collapse
Affiliation(s)
- Shane P. Simonett
- Biochemistry Department, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Sunyoung Shin
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Jacob A. Herring
- Nutrition, Dietetics and Food Science Department, Brigham Young University, Provo, Utah, USA
| | - Rhonda Bacher
- Department of Biostatistics, University of Florida, Gainesville, Florida, USA
| | - Linsin A. Smith
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, Illinois, USA
| | - Chenyang Dong
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Mary E. Rabaglia
- Biochemistry Department, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Donnie S. Stapleton
- Biochemistry Department, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Kathryn L. Schueler
- Biochemistry Department, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Jeea Choi
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | | | - Daniel R. Turkewitz
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Carlos Perez-Cervantes
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, Illinois, USA
| | - Jason Spaeth
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Roland Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Jeffery S. Tessem
- Nutrition, Dietetics and Food Science Department, Brigham Young University, Provo, Utah, USA
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Sündüz Keleş
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Ivan P. Moskowitz
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, Illinois, USA
| | - Mark P. Keller
- Biochemistry Department, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Alan D. Attie
- Biochemistry Department, University of Wisconsin–Madison, Madison, Wisconsin, USA
| |
Collapse
|
16
|
Kalwat MA, Scheuner D, Rodrigues-dos-Santos K, Eizirik DL, Cobb MH. The Pancreatic ß-cell Response to Secretory Demands and Adaption to Stress. Endocrinology 2021; 162:bqab173. [PMID: 34407177 PMCID: PMC8459449 DOI: 10.1210/endocr/bqab173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Indexed: 02/06/2023]
Abstract
Pancreatic β cells dedicate much of their protein translation capacity to producing insulin to maintain glucose homeostasis. In response to increased secretory demand, β cells can compensate by increasing insulin production capability even in the face of protracted peripheral insulin resistance. The ability to amplify insulin secretion in response to hyperglycemia is a critical facet of β-cell function, and the exact mechanisms by which this occurs have been studied for decades. To adapt to the constant and fast-changing demands for insulin production, β cells use the unfolded protein response of the endoplasmic reticulum. Failure of these compensatory mechanisms contributes to both type 1 and 2 diabetes. Additionally, studies in which β cells are "rested" by reducing endogenous insulin demand have shown promise as a therapeutic strategy that could be applied more broadly. Here, we review recent findings in β cells pertaining to the metabolic amplifying pathway, the unfolded protein response, and potential advances in therapeutics based on β-cell rest.
Collapse
Affiliation(s)
- Michael A Kalwat
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| | - Donalyn Scheuner
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| | | | - Decio L Eizirik
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Melanie H Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
17
|
Dong C, Simonett SP, Shin S, Stapleton DS, Schueler KL, Churchill GA, Lu L, Liu X, Jin F, Li Y, Attie AD, Keller MP, Keleş S. INFIMA leverages multi-omics model organism data to identify effector genes of human GWAS variants. Genome Biol 2021; 22:241. [PMID: 34425882 PMCID: PMC8381555 DOI: 10.1186/s13059-021-02450-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 08/02/2021] [Indexed: 11/24/2022] Open
Abstract
Genome-wide association studies reveal many non-coding variants associated with complex traits. However, model organism studies largely remain as an untapped resource for unveiling the effector genes of non-coding variants. We develop INFIMA, Integrative Fine-Mapping, to pinpoint causal SNPs for diversity outbred (DO) mice eQTL by integrating founder mice multi-omics data including ATAC-seq, RNA-seq, footprinting, and in silico mutation analysis. We demonstrate INFIMA's superior performance compared to alternatives with human and mouse chromatin conformation capture datasets. We apply INFIMA to identify novel effector genes for GWAS variants associated with diabetes. The results of the application are available at http://www.statlab.wisc.edu/shiny/INFIMA/ .
Collapse
Affiliation(s)
- Chenyang Dong
- Department of Statistics, University of Wisconsin-Madison, Madison, WI USA
| | - Shane P. Simonett
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI USA
| | - Sunyoung Shin
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, TX USA
| | - Donnie S. Stapleton
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI USA
| | - Kathryn L. Schueler
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI USA
| | | | - Leina Lu
- Case Western University, Cleveland, OH USA
| | | | - Fulai Jin
- Case Western University, Cleveland, OH USA
| | - Yan Li
- Case Western University, Cleveland, OH USA
| | - Alan D. Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI USA
| | - Mark P. Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI USA
| | - Sündüz Keleş
- Department of Statistics, University of Wisconsin-Madison, Madison, WI USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI USA
| |
Collapse
|
18
|
Yang GH, Fontaine DA, Lodh S, Blumer JT, Roopra A, Davis DB. TCF19 Impacts a Network of Inflammatory and DNA Damage Response Genes in the Pancreatic β-Cell. Metabolites 2021; 11:metabo11080513. [PMID: 34436454 PMCID: PMC8400192 DOI: 10.3390/metabo11080513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
Transcription factor 19 (TCF19) is a gene associated with type 1 diabetes (T1DM) and type 2 diabetes (T2DM) in genome-wide association studies. Prior studies have demonstrated that Tcf19 knockdown impairs β-cell proliferation and increases apoptosis. However, little is known about its role in diabetes pathogenesis or the effects of TCF19 gain-of-function. The aim of this study was to examine the impact of TCF19 overexpression in INS-1 β-cells and human islets on proliferation and gene expression. With TCF19 overexpression, there was an increase in nucleotide incorporation without any change in cell cycle gene expression, alluding to an alternate process of nucleotide incorporation. Analysis of RNA-seq of TCF19 overexpressing cells revealed increased expression of several DNA damage response (DDR) genes, as well as a tightly linked set of genes involved in viral responses, immune system processes, and inflammation. This connectivity between DNA damage and inflammatory gene expression has not been well studied in the β-cell and suggests a novel role for TCF19 in regulating these pathways. Future studies determining how TCF19 may modulate these pathways can provide potential targets for improving β-cell survival.
Collapse
Affiliation(s)
- Grace H. Yang
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
| | - Danielle A. Fontaine
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
| | - Sukanya Lodh
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53233, USA
| | - Joseph T. Blumer
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
| | - Avtar Roopra
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI 53705, USA;
| | - Dawn Belt Davis
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (G.H.Y.); (D.A.F.); (S.L.); (J.T.B.)
- William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Correspondence:
| |
Collapse
|
19
|
Oflaz FE, Koshenov Z, Hirtl M, Rost R, Bachkoenig OA, Gottschalk B, Madreiter-Sokolowski CT, Malli R, Graier WF. Near-UV Light Induced ROS Production Initiates Spatial Ca 2+ Spiking to Fire NFATc3 Translocation. Int J Mol Sci 2021; 22:8189. [PMID: 34360954 PMCID: PMC8346968 DOI: 10.3390/ijms22158189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 01/19/2023] Open
Abstract
Ca2+-dependent gene regulation controls several functions to determine the fate of the cells. Proteins of the nuclear factor of activated T-cells (NFAT) family are Ca2+ sensitive transcription factors that control the cell growth, proliferation and insulin secretion in β-cells. Translocation of NFAT proteins to the nucleus occurs in a sequence of events that starts with activating calmodulin-dependent phosphatase calcineurin in a Ca2+-dependent manner, which dephosphorylates the NFAT proteins and leads to their translocation to the nucleus. Here, we examined the role of IP3-generating agonists and near-UV light in the induction of NFATc3 migration to the nucleus in the pancreatic β-cell line INS-1. Our results show that IP3 generation yields cytosolic Ca2+ rise and NFATc3 translocation. Moreover, near-UV light exposure generates reactive oxygen species (ROS), resulting in cytosolic Ca2+ spiking via the L-type Ca2+ channel and triggers NFATc3 translocation to the nucleus. Using the mitochondria as a Ca2+ buffering tool, we showed that ROS-induced cytosolic Ca2+ spiking, not the ROS themselves, was the triggering mechanism of nuclear import of NFATc3. Collectively, this study reveals the mechanism of near-UV light induced NFATc3 migration.
Collapse
Affiliation(s)
- Furkan E. Oflaz
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Zhanat Koshenov
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Martin Hirtl
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Rene Rost
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Olaf A. Bachkoenig
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Benjamin Gottschalk
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Corina T. Madreiter-Sokolowski
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Roland Malli
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Wolfgang F. Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
20
|
Chan JY, Bensellam M, Lin RCY, Liang C, Lee K, Jonas JC, Laybutt DR. Transcriptome analysis of islets from diabetes-resistant and diabetes-prone obese mice reveals novel gene regulatory networks involved in beta-cell compensation and failure. FASEB J 2021; 35:e21608. [PMID: 33977593 DOI: 10.1096/fj.202100009r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/23/2021] [Accepted: 04/05/2021] [Indexed: 01/02/2023]
Abstract
The mechanisms underpinning beta-cell compensation for obesity-associated insulin resistance and beta-cell failure in type 2 diabetes remain poorly understood. We used a large-scale strategy to determine the time-dependent transcriptomic changes in islets of diabetes-prone db/db and diabetes-resistant ob/ob mice at 6 and 16 weeks of age. Differentially expressed genes were subjected to cluster, gene ontology, pathway and gene set enrichment analyses. A distinctive gene expression pattern was observed in 16 week db/db islets in comparison to the other groups with alterations in transcriptional regulators of islet cell identity, upregulation of glucose/lipid metabolism, and various stress response genes, and downregulation of specific amino acid transport and metabolism genes. In contrast, ob/ob islets displayed a coordinated downregulation of metabolic and stress response genes at 6 weeks of age, suggestive of a preemptive reconfiguration in these islets to lower the threshold of metabolic activation in response to increased insulin demand thereby preserving beta-cell function and preventing cellular stress. In addition, amino acid transport and metabolism genes were upregulated in ob/ob islets, suggesting an important role of glutamate metabolism in beta-cell compensation. Gene set enrichment analysis of differentially expressed genes identified the enrichment of binding motifs for transcription factors, FOXO4, NFATC1, and MAZ. siRNA-mediated knockdown of these genes in MIN6 cells altered cell death, insulin secretion, and stress gene expression. In conclusion, these data revealed novel gene regulatory networks involved in beta-cell compensation and failure. Preemptive metabolic reconfiguration in diabetes-resistant islets may dampen metabolic activation and cellular stress during obesity.
Collapse
Affiliation(s)
- Jeng Yie Chan
- Garvan Institute of Medical Research, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Mohammed Bensellam
- Garvan Institute of Medical Research, Sydney, NSW, Australia.,Pôle D'endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Ruby C Y Lin
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.,Centre for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Sydney, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Cassandra Liang
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Kailun Lee
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Jean-Christophe Jonas
- Pôle D'endocrinologie, Diabète et Nutrition, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - D Ross Laybutt
- Garvan Institute of Medical Research, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
21
|
Wickramasinghe ASD, Kalansuriya P, Attanayake AP. Herbal Medicines Targeting the Improved β-Cell Functions and β-Cell Regeneration for the Management of Diabetes Mellitus. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:2920530. [PMID: 34335803 PMCID: PMC8298154 DOI: 10.1155/2021/2920530] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022]
Abstract
There is an increasing trend of investigating natural bioactive compounds targeting pancreatic β-cells for the prevention/treatment of diabetes mellitus (DM). With the exploration of multiple mechanisms by which β-cells involve in the pathogenesis of DM, herbal medicines are gaining attention due to their multitasking ability as evidenced by traditional medicine practices. This review attempts to summarize herbal medicines with the potential for improvement of β-cell functions and regeneration as scientifically proven by in vivo/in vitro investigations. Furthermore, attempts have been made to identify the mechanisms of improving the function and regeneration of β-cells by herbal medicines. Relevant data published from January 2009 to March 2020 were collected by searching electronic databases "PubMed," "ScienceDirect," and "Google Scholar" and studied for this review. Single herbal extracts, polyherbal mixtures, and isolated compounds derived from approximately 110 medicinal plants belonging to 51 different plant families had been investigated in recent years and found to be targeting β-cells. Many herbal medicines showed improvement of β-cell function as observed through homeostatic model assessment-β-cell function (HOMA-β). Pancreatic β-cell regeneration as observed in histopathological and immunohistochemical studies in terms of increase of size and number of functional β-cells was also prominent. Increasing β-cell mass via expression of genes/proteins related to antiapoptotic actions and β-cell neogenesis/proliferation, increasing glucose-stimulated insulin secretion via activating glucose transporter-2 (GLUT-2) receptors, and/or increasing intracellular Ca2+ levels were observed upon treatment of some herbal medicines. Some herbal medicines acted on various insulin signaling pathways. Furthermore, many herbal medicines showed protective effects on β-cells via reduction of oxidative stress and inflammation. However, there are many unexplored avenues. Thus, further investigations are warranted in elucidating mechanisms of improving β-cell function and mass by herbal medicines, their structure-activity relationship (SAR), and toxicities of these herbal medicines.
Collapse
Affiliation(s)
| | - Pabasara Kalansuriya
- Department of Biochemistry, Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
| | | |
Collapse
|
22
|
Wortham M, Sander M. Transcriptional mechanisms of pancreatic β-cell maturation and functional adaptation. Trends Endocrinol Metab 2021; 32:474-487. [PMID: 34030925 PMCID: PMC8259463 DOI: 10.1016/j.tem.2021.04.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 12/31/2022]
Abstract
Pancreatic β-cells secrete insulin commensurate to circulating nutrient levels to maintain normoglycemia. The ability of β-cells to couple insulin secretion to nutrient stimuli is acquired during a postnatal maturation process. In mature β-cells the insulin secretory response adapts to changes in nutrient state. Both β-cell maturation and functional adaptation rely on the interplay between extracellular cues and cell type-specific transcriptional programs. Here we review emerging evidence that developmental and homeostatic regulation of β-cell function involves collaboration between lineage-determining and signal-dependent transcription factors (LDTFs and SDTFs, respectively). A deeper understanding of β-cell SDTFs and their cognate signals would delineate mechanisms of β-cell maturation and functional adaptation, which has direct implications for diabetes therapies and for generating mature β-cells from stem cells.
Collapse
Affiliation(s)
- Matthew Wortham
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Maike Sander
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
23
|
Karakose E, Wang H, Inabnet W, Thakker RV, Libutti S, Fernandez-Ranvier G, Suh H, Stevenson M, Kinoshita Y, Donovan M, Antipin Y, Li Y, Liu X, Jin F, Wang P, Uzilov A, Argmann C, Schadt EE, Stewart AF, Scott DK, Lambertini L. Aberrant methylation underlies insulin gene expression in human insulinoma. Nat Commun 2020; 11:5210. [PMID: 33060578 PMCID: PMC7566641 DOI: 10.1038/s41467-020-18839-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 09/16/2020] [Indexed: 12/23/2022] Open
Abstract
Human insulinomas are rare, benign, slowly proliferating, insulin-producing beta cell tumors that provide a molecular "recipe" or "roadmap" for pathways that control human beta cell regeneration. An earlier study revealed abnormal methylation in the imprinted p15.5-p15.4 region of chromosome 11, known to be abnormally methylated in another disorder of expanded beta cell mass and function: the focal variant of congenital hyperinsulinism. Here, we compare deep DNA methylome sequencing on 19 human insulinomas, and five sets of normal beta cells. We find a remarkably consistent, abnormal methylation pattern in insulinomas. The findings suggest that abnormal insulin (INS) promoter methylation and altered transcription factor expression create alternative drivers of INS expression, replacing canonical PDX1-driven beta cell specification with a pathological, looping, distal enhancer-based form of transcriptional regulation. Finally, NFaT transcription factors, rather than the canonical PDX1 enhancer complex, are predicted to drive INS transactivation.
Collapse
Affiliation(s)
- Esra Karakose
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - William Inabnet
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rajesh V Thakker
- The Academic Endocrine Unit, University of Oxford, OX3 7LJ, Oxford, UK
| | - Steven Libutti
- The Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA
| | - Gustavo Fernandez-Ranvier
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Hyunsuk Suh
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mark Stevenson
- The Academic Endocrine Unit, University of Oxford, OX3 7LJ, Oxford, UK
| | - Yayoi Kinoshita
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Michael Donovan
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yevgeniy Antipin
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Sema4, Stamford, CT, 06902, USA
| | - Yan Li
- The Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Xiaoxiao Liu
- The Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Fulai Jin
- The Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Peng Wang
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Andrew Uzilov
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Sema4, Stamford, CT, 06902, USA
| | - Carmen Argmann
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric E Schadt
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Sema4, Stamford, CT, 06902, USA
| | - Andrew F Stewart
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Donald K Scott
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Luca Lambertini
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
24
|
CDK8 Regulates Insulin Secretion and Mediates Postnatal and Stress-Induced Expression of Neuropeptides in Pancreatic β Cells. Cell Rep 2020; 28:2892-2904.e7. [PMID: 31509750 DOI: 10.1016/j.celrep.2019.08.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/26/2019] [Accepted: 08/05/2019] [Indexed: 12/13/2022] Open
Abstract
Cyclin-dependent kinases (CDKs) contribute to vital cellular processes including cell cycle regulation. Loss of CDKs is associated with impaired insulin secretion and β cell survival; however, the function of CDK8 in β cells remains elusive. Here, we report that genetic ablation of Cdk8 improves glucose tolerance by increasing insulin secretion. We identify OSBPL3 as a CDK8-dependent phosphoprotein, which acts as a negative regulator of insulin secretion in response to glucose. We also show that embryonic gene silencing of neuropeptide Y in β cells is compromised in Cdk8-null mice, leading to continued expression into adulthood. Cdk8 ablation in β cells aggravates apoptosis and induces de novo expression of neuropeptides upon oxidative stress. Moreover, pancreatic islets exposed to stress display augmented apoptosis in the presence of these same neuropeptides. Our results reveal critical roles for CDK8 in β cell function and survival during metabolic stress that are in part mediated through de novo expression of neuropeptides.
Collapse
|
25
|
Mouse Systems Genetics as a Prelude to Precision Medicine. Trends Genet 2020; 36:259-272. [PMID: 32037011 DOI: 10.1016/j.tig.2020.01.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 12/17/2022]
Abstract
Mouse models have been instrumental in understanding human disease biology and proposing possible new treatments. The precise control of the environment and genetic composition of mice allows more rigorous observations, but limits the generalizability and translatability of the results into human applications. In the era of precision medicine, strategies using mouse models have to be revisited to effectively emulate human populations. Systems genetics is one promising paradigm that may promote the transition to novel precision medicine strategies. Here, we review the state-of-the-art resources and discuss how mouse systems genetics helps to understand human diseases and to advance the development of precision medicine, with an emphasis on the existing resources and strategies.
Collapse
|
26
|
Hu L, He F, Luo Y, Luo H, Hai L, Li Y, Zhou Z, Liu F, Dai YS. Reduced Compensatory β-Cell Proliferation in Nfatc3-Deficient Mice Fed on High-Fat Diet. Exp Clin Endocrinol Diabetes 2019; 129:651-660. [PMID: 31546271 DOI: 10.1055/a-1008-9110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND High-fat-diet induces pancreatic β-cell compensatory proliferation, and impairments in pancreatic β-cell proliferation and function can lead to defects in insulin secretion and diabetes. NFATc3 is important for HFD-induced adipose tissue inflammation. But it is unknown whether NFATc3 is required for β cell compensatory growth in mice fed with HFD. METHODS NFATc3 mRNA and protein expression levels were quantified by RT-qPCR and Western blotting, respectively, in pancreatic islets of WT mice fed on HFD for 12-20 weeks. Adenoviral-mediated overexpression of NFATc3 were conducted in Min6 cells and cultured primary mouse islets. NFATc3-/- mice and WT control mice were fed with HFD and metabolic and functional parameters were measured. RESULTS We observed that the NFATc3 expression level was reduced in the islets of high-fat-diet (HFD)-fed mice. Adenovirus-mediated overexpression of NFATc3 enhanced glucose-stimulated insulin secretion and β-cell gene expression in cultured primary mouse islets. Nfatc3-/- mice initially developed similar glucose tolerance at 2-4 weeks after HFD feeding than HFD-fed WT mice, but Nfatc3-/- mice developed improved glucose tolerance and insulin sensitivity after 8 weeks of HFD feeding compared to Nfatc3+/+fed with HFD. Furthermore, Nfatc3-/- mice on HFD exhibited decreased β-cell mass and reduced expression of genes important for β-cell proliferation and function compared to Nfatc3+/+mice on HFD. CONCLUSIONS The findings suggested that NFATc3 plays a role in maintaining the pancreatic β-cell compensatory growth and gene expression in response to obesity.
Collapse
Affiliation(s)
- Li Hu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fengli He
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Luo
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hairong Luo
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luo Hai
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yabin Li
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Liu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Yan-Shan Dai
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Bristol-Myers Squibb Company, Princeton, NJ 08540, United States
| |
Collapse
|
27
|
Identification of C2CD4A as a human diabetes susceptibility gene with a role in β cell insulin secretion. Proc Natl Acad Sci U S A 2019; 116:20033-20042. [PMID: 31527256 DOI: 10.1073/pnas.1904311116] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Fine mapping and validation of genes causing β cell failure from susceptibility loci identified in type 2 diabetes genome-wide association studies (GWAS) poses a significant challenge. The VPS13C-C2CD4A-C2CD4B locus on chromosome 15 confers diabetes susceptibility in every ethnic group studied to date. However, the causative gene is unknown. FoxO1 is involved in the pathogenesis of β cell dysfunction, but its link to human diabetes GWAS has not been explored. Here we generated a genome-wide map of FoxO1 superenhancers in chemically identified β cells using 2-photon live-cell imaging to monitor FoxO1 localization. When parsed against human superenhancers and GWAS-derived diabetes susceptibility alleles, this map revealed a conserved superenhancer in C2CD4A, a gene encoding a β cell/stomach-enriched nuclear protein of unknown function. Genetic ablation of C2cd4a in β cells of mice phenocopied the metabolic abnormalities of human carriers of C2CD4A-linked polymorphisms, resulting in impaired insulin secretion during glucose tolerance tests as well as hyperglycemic clamps. C2CD4A regulates glycolytic genes, and notably represses key β cell "disallowed" genes, such as lactate dehydrogenase A We propose that C2CD4A is a transcriptional coregulator of the glycolytic pathway whose dysfunction accounts for the diabetes susceptibility associated with the chromosome 15 GWAS locus.
Collapse
|
28
|
Sharp-Tawfik AE, Coiner AM, MarElia CB, Kazantzis M, Zhang C, Burkhardt BR. Compositional analysis and biological characterization of Cornus officinalis on human 1.1B4 pancreatic β cells. Mol Cell Endocrinol 2019; 494:110491. [PMID: 31255730 DOI: 10.1016/j.mce.2019.110491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/21/2019] [Accepted: 06/22/2019] [Indexed: 12/16/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease resulting from the loss of pancreatic β cells and subsequent insulin production. Novel interventional therapies are urgently needed that can protect existing β cells from cytokine-induced death and enhance their function before symptomatic onset. Our initial evidence is suggesting that bioactive ingredients within Cornus officinalis (CO) may be able to serve in this function. CO has been extensively used in Traditional Chinese Medicine (TCM) and reported to possess both anti-inflammatory and pro-metabolic effects. We hypothesize that CO treatment may provide a future potential candidate for interventional therapy for early stage T1D prior to significant β cell loss. Our data demonstrated that CO can inhibit cytokine-mediated β cell death, increase cell viability and oxidative capacity, and increase expression of NFATC2 (Nuclear Factor of Activated T Cells, Cytoplasmic 2). We have also profiled the bioactive components in CO from multiple sources by HPLC/MS (High Performance Liquid Chromatography/Mass Spectrometry) analysis. Altogether, CO significantly increases the energy metabolism of β cells while inducing the NFAT pathway to signal for increased proliferation and endocrine function.
Collapse
Affiliation(s)
- Arielle E Sharp-Tawfik
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA
| | - Alexis M Coiner
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA
| | - Catherine B MarElia
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA
| | - Melissa Kazantzis
- Metabolic Core, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Clare Zhang
- Practice of Oriental Medicine, Tucson, AZ, USA
| | - Brant R Burkhardt
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA.
| |
Collapse
|
29
|
Transcriptome profiling reveals the anti-diabetic molecular mechanism of Cyclocarya paliurus polysaccharides. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.12.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
30
|
Sabatini PV, Speckmann T, Lynn FC. Friend and foe: β-cell Ca 2+ signaling and the development of diabetes. Mol Metab 2019; 21:1-12. [PMID: 30630689 PMCID: PMC6407368 DOI: 10.1016/j.molmet.2018.12.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/03/2018] [Accepted: 12/19/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The divalent cation Calcium (Ca2+) regulates a wide range of processes in disparate cell types. Within insulin-producing β-cells, increases in cytosolic Ca2+ directly stimulate insulin vesicle exocytosis, but also initiate multiple signaling pathways. Mediated through activation of downstream kinases and transcription factors, Ca2+-regulated signaling pathways leverage substantial influence on a number of critical cellular processes within the β-cell. Additionally, there is evidence that prolonged activation of these same pathways is detrimental to β-cell health and may contribute to Type 2 Diabetes pathogenesis. SCOPE OF REVIEW This review aims to briefly highlight canonical Ca2+ signaling pathways in β-cells and how β-cells regulate the movement of Ca2+ across numerous organelles and microdomains. As a main focus, this review synthesizes experimental data from in vitro and in vivo models on both the beneficial and detrimental effects of Ca2+ signaling pathways for β-cell function and health. MAJOR CONCLUSIONS Acute increases in intracellular Ca2+ stimulate a number of signaling cascades, resulting in (de-)phosphorylation events and activation of downstream transcription factors. The short-term stimulation of these Ca2+ signaling pathways promotes numerous cellular processes critical to β-cell function, including increased viability, replication, and insulin production and secretion. Conversely, chronic stimulation of Ca2+ signaling pathways increases β-cell ER stress and results in the loss of β-cell differentiation status. Together, decades of study demonstrate that Ca2+ movement is tightly regulated within the β-cell, which is at least partially due to its dual roles as a potent signaling molecule.
Collapse
Affiliation(s)
- Paul V Sabatini
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Thilo Speckmann
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Francis C Lynn
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
31
|
Kycia I, Wolford BN, Huyghe JR, Fuchsberger C, Vadlamudi S, Kursawe R, Welch RP, Albanus RD, Uyar A, Khetan S, Lawlor N, Bolisetty M, Mathur A, Kuusisto J, Laakso M, Ucar D, Mohlke KL, Boehnke M, Collins FS, Parker SCJ, Stitzel ML. A Common Type 2 Diabetes Risk Variant Potentiates Activity of an Evolutionarily Conserved Islet Stretch Enhancer and Increases C2CD4A and C2CD4B Expression. Am J Hum Genet 2018; 102:620-635. [PMID: 29625024 DOI: 10.1016/j.ajhg.2018.02.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 02/22/2018] [Indexed: 01/17/2023] Open
Abstract
Genome-wide association studies (GWASs) and functional genomics approaches implicate enhancer disruption in islet dysfunction and type 2 diabetes (T2D) risk. We applied genetic fine-mapping and functional (epi)genomic approaches to a T2D- and proinsulin-associated 15q22.2 locus to identify a most likely causal variant, determine its direction of effect, and elucidate plausible target genes. Fine-mapping and conditional analyses of proinsulin levels of 8,635 non-diabetic individuals from the METSIM study support a single association signal represented by a cluster of 16 strongly associated (p < 10-17) variants in high linkage disequilibrium (r2 > 0.8) with the GWAS index SNP rs7172432. These variants reside in an evolutionarily and functionally conserved islet and β cell stretch or super enhancer; the most strongly associated variant (rs7163757, p = 3 × 10-19) overlaps a conserved islet open chromatin site. DNA sequence containing the rs7163757 risk allele displayed 2-fold higher enhancer activity than the non-risk allele in reporter assays (p < 0.01) and was differentially bound by β cell nuclear extract proteins. Transcription factor NFAT specifically potentiated risk-allele enhancer activity and altered patterns of nuclear protein binding to the risk allele in vitro, suggesting that it could be a factor mediating risk-allele effects. Finally, the rs7163757 proinsulin-raising and T2D risk allele (C) was associated with increased expression of C2CD4B, and possibly C2CD4A, both of which were induced by inflammatory cytokines, in human islets. Together, these data suggest that rs7163757 contributes to genetic risk of islet dysfunction and T2D by increasing NFAT-mediated islet enhancer activity and modulating C2CD4B, and possibly C2CD4A, expression in (patho)physiologic states.
Collapse
Affiliation(s)
- Ina Kycia
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Brooke N Wolford
- National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Jeroen R Huyghe
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Christian Fuchsberger
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Romy Kursawe
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Ryan P Welch
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ricardo d'Oliveira Albanus
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Asli Uyar
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Shubham Khetan
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Nathan Lawlor
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Mohan Bolisetty
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Anubhuti Mathur
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Duygu Ucar
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Francis S Collins
- National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Stephen C J Parker
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael L Stitzel
- Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut Health Center, Farmington, CT 06032, USA.
| |
Collapse
|
32
|
Ye R, Gordillo R, Shao M, Onodera T, Chen Z, Chen S, Lin X, SoRelle JA, Li X, Tang M, Keller MP, Kuliawat R, Attie AD, Gupta RK, Holland WL, Beutler B, Herz J, Scherer PE. Intracellular lipid metabolism impairs β cell compensation during diet-induced obesity. J Clin Invest 2018; 128:1178-1189. [PMID: 29457786 DOI: 10.1172/jci97702] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/09/2018] [Indexed: 12/16/2022] Open
Abstract
The compensatory proliferation of insulin-producing β cells is critical to maintaining glucose homeostasis at the early stage of type 2 diabetes. Failure of β cells to proliferate results in hyperglycemia and insulin dependence in patients. To understand the effect of the interplay between β cell compensation and lipid metabolism upon obesity and peripheral insulin resistance, we eliminated LDL receptor-related protein 1 (LRP1), a pleiotropic mediator of cholesterol, insulin, energy metabolism, and other cellular processes, in β cells. Upon high-fat diet exposure, LRP1 ablation significantly impaired insulin secretion and proliferation of β cells. The diminished insulin signaling was partly contributed to by the hypersensitivity to glucose-induced, Ca2+-dependent activation of Erk and the mTORC1 effector p85 S6K1. Surprisingly, in LRP1-deficient islets, lipotoxic sphingolipids were mitigated by improved lipid metabolism, mediated at least in part by the master transcriptional regulator PPARγ2. Acute overexpression of PPARγ2 in β cells impaired insulin signaling and insulin secretion. Elimination of Apbb2, a functional regulator of LRP1 cytoplasmic domain, also impaired β cell function in a similar fashion. In summary, our results uncover the double-edged effects of intracellular lipid metabolism on β cell function and viability in obesity and type 2 diabetes and highlight LRP1 as an essential regulator of these processes.
Collapse
Affiliation(s)
- Risheng Ye
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern (UTSW) Medical Center, Dallas, Texas, USA.,Department of Medical Education, Texas Tech University Health Sciences Center Paul L. Foster School of Medicine, El Paso, Texas, USA
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern (UTSW) Medical Center, Dallas, Texas, USA
| | - Mengle Shao
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern (UTSW) Medical Center, Dallas, Texas, USA
| | - Toshiharu Onodera
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern (UTSW) Medical Center, Dallas, Texas, USA
| | - Zhe Chen
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern (UTSW) Medical Center, Dallas, Texas, USA.,Center for the Genetics of Host Defense, UTSW Medical Center, Dallas, Texas, USA
| | - Shiuhwei Chen
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern (UTSW) Medical Center, Dallas, Texas, USA
| | - Xiaoli Lin
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern (UTSW) Medical Center, Dallas, Texas, USA
| | - Jeffrey A SoRelle
- Center for the Genetics of Host Defense, UTSW Medical Center, Dallas, Texas, USA
| | - Xiaohong Li
- Center for the Genetics of Host Defense, UTSW Medical Center, Dallas, Texas, USA
| | - Miao Tang
- Center for the Genetics of Host Defense, UTSW Medical Center, Dallas, Texas, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin, USA
| | - Regina Kuliawat
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, New York, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin, Madison, Wisconsin, USA
| | - Rana K Gupta
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern (UTSW) Medical Center, Dallas, Texas, USA
| | - William L Holland
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern (UTSW) Medical Center, Dallas, Texas, USA
| | - Bruce Beutler
- Center for the Genetics of Host Defense, UTSW Medical Center, Dallas, Texas, USA
| | - Joachim Herz
- Departments of Molecular Genetics, Neuroscience, Neurology and Neurotherapeutics, and Center for Translational Neurodegeneration Research, UTSW Medical Center, Dallas, Texas, USA.,Center for Neuroscience, Department of Neuroanatomy, Albert Ludwig University, Freiburg, Germany
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern (UTSW) Medical Center, Dallas, Texas, USA
| |
Collapse
|
33
|
Lawlor N, Khetan S, Ucar D, Stitzel ML. Genomics of Islet (Dys)function and Type 2 Diabetes. Trends Genet 2017; 33:244-255. [PMID: 28245910 DOI: 10.1016/j.tig.2017.01.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 01/30/2017] [Indexed: 12/28/2022]
Abstract
Pancreatic islet dysfunction and beta cell failure are hallmarks of type 2 diabetes mellitus (T2DM) pathogenesis. In this review, we discuss how genome-wide association studies (GWASs) and recent developments in islet (epi)genome and transcriptome profiling (particularly single cell analyses) are providing novel insights into the genetic, environmental, and cellular contributions to islet (dys)function and T2DM pathogenesis. Moving forward, study designs that interrogate and model genetic variation [e.g., allelic profiling and (epi)genome editing] will be critical to dissect the molecular genetics of T2DM pathogenesis, to build next-generation cellular and animal models, and to develop precision medicine approaches to detect, treat, and prevent islet (dys)function and T2DM.
Collapse
Affiliation(s)
- Nathan Lawlor
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Shubham Khetan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics & Genome Sciences, University of Connecticut, Farmington, CT 06032, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Institute for Systems Genomics, University of Connecticut, Farmington, CT 06032, USA
| | - Michael L Stitzel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics & Genome Sciences, University of Connecticut, Farmington, CT 06032, USA; Institute for Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| |
Collapse
|