1
|
Yang J, Zhou Y, Zhang J, Zheng Y, He J. Identification of genes related to fatty acid metabolism in type 2 diabetes mellitus. Biochem Biophys Rep 2024; 40:101849. [PMID: 39498440 PMCID: PMC11532806 DOI: 10.1016/j.bbrep.2024.101849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 11/07/2024] Open
Abstract
Aim Fatty acid metabolism is pivotal for lipid synthesis, cellular signaling, and maintaining cell membrane integrity. However, its diagnostic significance in type 2 diabetes mellitus (T2DM) remains unclear. Materials and methods Three datasets and fatty acid metabolism-related genes were retrieved. Differential expression analysis, WGCNA, machine learning algorithms, diagnostic analysis, and validation were employed to identify key feature genes. Functional analysis, ceRNA network construction, immune microenvironment assessment, and drug prediction were conducted to explore the underlying molecular mechanisms. Results Six feature genes were identified with strong diagnostic performance and were involved in processes such as ribosome function and fatty acid metabolism. Immune cells, including dendritic cells, eosinophils, and neutrophils, may play a role in the progression of T2DM. ceRNA and drug-target network analysis revealed potential interactions, such as RP11-miR-29a-YTHDF3 and BPA-MSANTD1. The expression patterns of the feature genes, except for YTHDF3, were consistently upregulated in T2DM, aligning with trends observed in the training set. Conclusion This study investigated the potential molecular mechanisms of six fatty acid metabolism-related genes in T2DM, offering valuable insights that may guide future research and therapeutic development.
Collapse
Affiliation(s)
- Ji Yang
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
- Department of Endocrinology and Metabolism, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yikun Zhou
- Department of Endocrinology and Metabolism, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jiarui Zhang
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
- Department of Endocrinology and Metabolism, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yongqin Zheng
- Department of Endocrinology and Metabolism, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jundong He
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
- Department of Endocrinology and Metabolism, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
2
|
Lien YC, Pinney SE, Lu XM, Simmons RA. Identification of Novel Regulatory Regions Induced by Intrauterine Growth Restriction in Rat Islets. Endocrinology 2022; 163:6459683. [PMID: 34894232 PMCID: PMC8743043 DOI: 10.1210/endocr/bqab251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Indexed: 01/05/2023]
Abstract
Intrauterine growth restriction (IUGR) leads to the development of type 2 diabetes in adulthood, and the permanent alterations in gene expression implicate an epigenetic mechanism. Using a rat model of IUGR, we performed TrueSeq-HELP Tagging to assess the association of DNA methylation changes and gene dysregulation in islets. We identified 511 differentially methylated regions (DMRs) and 4377 significantly altered single CpG sites. Integrating the methylome and our published transcriptome data sets resulted in the identification of pathways critical for islet function. The identified DMRs were enriched with transcription factor binding motifs, such as Elk1, Etv1, Foxa1, Foxa2, Pax7, Stat3, Hnf1, and AR. In silico analysis of 3-dimensional chromosomal interactions using human pancreas and islet Hi-C data sets identified interactions between 14 highly conserved DMRs and 35 genes with significant expression changes at an early age, many of which persisted in adult islets. In adult islets, there were far more interactions between DMRs and genes with significant expression changes identified with Hi-C, and most of them were critical to islet metabolism and insulin secretion. The methylome was integrated with our published genome-wide histone modification data sets from IUGR islets, resulting in further characterization of important regulatory regions of the genome altered by IUGR containing both significant changes in DNA methylation and specific histone marks. We identified novel regulatory regions in islets after exposure to IUGR, suggesting that epigenetic changes at key transcription factor binding motifs and other gene regulatory regions may contribute to gene dysregulation and an abnormal islet phenotype in IUGR rats.
Collapse
Affiliation(s)
- Yu-Chin Lien
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Sara E Pinney
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Division Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Xueqing Maggie Lu
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Rebecca A Simmons
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
- Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Correspondence: Rebecca A. Simmons, MD, Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, the University of Pennsylvania, BRB II/III, 13th Fl, Rm 1308, 421 Curie Blvd, Philadelphia, PA 19104, USA.
| |
Collapse
|
3
|
Johnson CSC, Shively C, Michalson KT, Lea AJ, DeBo RJ, Howard TD, Hawkins GA, Appt SE, Liu Y, McCall CE, Herrington DM, Ip EH, Register TC, Snyder-Mackler N. Contrasting effects of Western vs Mediterranean diets on monocyte inflammatory gene expression and social behavior in a primate model. eLife 2021; 10:68293. [PMID: 34338633 PMCID: PMC8423447 DOI: 10.7554/elife.68293] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/28/2021] [Indexed: 01/20/2023] Open
Abstract
Dietary changes associated with industrialization increase the prevalence of chronic diseases, such as obesity, type II diabetes, and cardiovascular disease. This relationship is often attributed to an 'evolutionary mismatch' between human physiology and modern nutritional environments. Western diets enriched with foods that were scarce throughout human evolutionary history (e.g. simple sugars and saturated fats) promote inflammation and disease relative to diets more akin to ancestral human hunter-gatherer diets, such as a Mediterranean diet. Peripheral blood monocytes, precursors to macrophages and important mediators of innate immunity and inflammation, are sensitive to the environment and may represent a critical intermediate in the pathway linking diet to disease. We evaluated the effects of 15 months of whole diet manipulations mimicking Western or Mediterranean diet patterns on monocyte polarization in a well-established model of human health, the cynomolgus macaque (Macaca fascicularis). Monocyte transcriptional profiles differed markedly between diets, with 40% of transcripts showing differential expression (FDR < 0.05). Monocytes from Western diet consumers were polarized toward a more proinflammatory phenotype. The Western diet shifted the co-expression of 445 gene pairs, including small RNAs and transcription factors associated with metabolism and adiposity in humans, and dramatically altered behavior. For example, Western-fed individuals were more anxious and less socially integrated. These behavioral changes were also associated with some of the effects of diet on gene expression, suggesting an interaction between diet, central nervous system activity, and monocyte gene expression. This study provides new molecular insights into an evolutionary mismatch and uncovers new pathways through which Western diets alter monocyte polarization toward a proinflammatory phenotype.
Collapse
Affiliation(s)
- Corbin SC Johnson
- Department of Psychology, University of WashingtonSeattleUnited States
| | - Carol Shively
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of MedicineWinston-SalemUnited States
| | - Kristofer T Michalson
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of MedicineWinston-SalemUnited States
| | - Amanda J Lea
- Lewis-Sigler Institute for Integrative Genomics, Princeton UniversityPrincetonUnited States,Department of Ecology and Evolutionary Biology, Princeton UniversityPrincetonUnited States
| | - Ryne J DeBo
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of MedicineWinston-SalemUnited States
| | - Timothy D Howard
- Department of Biochemistry, Wake Forest School of MedicineWinston-SalemUnited States
| | - Gregory A Hawkins
- Department of Biochemistry, Wake Forest School of MedicineWinston-SalemUnited States
| | - Susan E Appt
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of MedicineWinston-SalemUnited States
| | - Yongmei Liu
- Division of Cardiology, Duke University School of MedicineDurhamUnited States
| | - Charles E McCall
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of MedicineWinston-SalemUnited States
| | - David M Herrington
- Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of MedicineWinston-SalemUnited States
| | - Edward H Ip
- Department of Biostatistics and Data Science, Wake Forest School of MedicineWinston-SalemUnited States
| | - Thomas C Register
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of MedicineWinston-SalemUnited States
| | - Noah Snyder-Mackler
- Department of Psychology, University of WashingtonSeattleUnited States,Center for Studies in Demography and Ecology, University of WashingtonSeattleUnited States,Department of Biology, University of WashingtonSeattleUnited States,School of Life Sciences, Arizona State UniversityTempeUnited States,Center for Evolution & Medicine, Arizona State UniversityTempeUnited States
| |
Collapse
|
4
|
Cao Y, Li L, Xu M, Feng Z, Sun X, Lu J, Xu Y, Du P, Wang T, Hu R, Ye Z, Shi L, Tang X, Yan L, Gao Z, Chen G, Zhang Y, Chen L, Ning G, Bi Y, Wang W. The ChinaMAP analytics of deep whole genome sequences in 10,588 individuals. Cell Res 2020; 30:717-731. [PMID: 32355288 PMCID: PMC7609296 DOI: 10.1038/s41422-020-0322-9] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/09/2020] [Indexed: 12/12/2022] Open
Abstract
Metabolic diseases are the most common and rapidly growing health issues worldwide. The massive population-based human genetics is crucial for the precise prevention and intervention of metabolic disorders. The China Metabolic Analytics Project (ChinaMAP) is based on cohort studies across diverse regions and ethnic groups with metabolic phenotypic data in China. Here, we describe the centralized analysis of the deep whole genome sequencing data and the genetic bases of metabolic traits in 10,588 individuals from the ChinaMAP. The frequency spectrum of variants, population structure, pathogenic variants and novel genomic characteristics were analyzed. The individual genetic evaluations of Mendelian diseases, nutrition and drug metabolism, and traits of blood glucose and BMI were integrated. Our study establishes a large-scale and deep resource for the genetics of East Asians and provides opportunities for novel genetic discoveries of metabolic characteristics and disorders.
Collapse
Affiliation(s)
- Yanan Cao
- National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- National Research Center for Translational Medicine, National Key Scientific Infrastructure for Translational Medicine (Shanghai), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lin Li
- National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- National Research Center for Translational Medicine, National Key Scientific Infrastructure for Translational Medicine (Shanghai), Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Min Xu
- National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhimin Feng
- National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaohui Sun
- National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jieli Lu
- National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu Xu
- National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Peina Du
- National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tiange Wang
- National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ruying Hu
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, 310006, Zhejiang, China
| | - Zhen Ye
- Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, 310006, Zhejiang, China
| | - Lixin Shi
- Affiliated Hospital of Guiyang Medical College, Guiyang, 550004, Guizhou, China
| | - Xulei Tang
- The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Li Yan
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Zhengnan Gao
- Dalian Municipal Central Hospital, Dalian, 116033, Liaoning, China
| | - Gang Chen
- Fujian Provincial Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Yinfei Zhang
- Central Hospital of Shanghai Jiading District, Shanghai, 201800, China
| | - Lulu Chen
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Guang Ning
- National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Yufang Bi
- National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Weiqing Wang
- National Clinical Research Centre for Metabolic Diseases, State Key Laboratory of Medical Genomics, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Institute for Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
5
|
Krentz NAJ, Gloyn AL. Insights into pancreatic islet cell dysfunction from type 2 diabetes mellitus genetics. Nat Rev Endocrinol 2020; 16:202-212. [PMID: 32099086 DOI: 10.1038/s41574-020-0325-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/17/2020] [Indexed: 12/30/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is an increasingly prevalent multifactorial disease that has both genetic and environmental risk factors, resulting in impaired glucose homeostasis. Genome-wide association studies (GWAS) have identified over 400 genetic signals that are associated with altered risk of T2DM. Human physiology and epigenomic data support a central role for the pancreatic islet in the pathogenesis of T2DM. This Review focuses on the promises and challenges of moving from genetic associations to molecular mechanisms and highlights efforts to identify the causal variant and effector transcripts at T2DM GWAS susceptibility loci. In addition, we examine current human models that are used to study both β-cell development and function, including EndoC-β cell lines and human induced pluripotent stem cell-derived β-like cells. We use examples of four T2DM susceptibility loci (CDKAL1, MTNR1B, SLC30A8 and PAM) to emphasize how a holistic approach involving genetics, physiology, and cellular and developmental biology can disentangle disease mechanisms at T2DM GWAS signals.
Collapse
Affiliation(s)
- Nicole A J Krentz
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
| | - Anna L Gloyn
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Churchill Hospital, Oxford, UK.
- Stanford Diabetes Research Centre, Stanford University, Stanford, CA, USA.
| |
Collapse
|
6
|
Jiao Y, Zhao J, Zhang Z, Li M, Yu X, Yang Y, Liu J, Liao S, Li D, Wang Y, Zhang D, Chen Y, Shi G, Liu B, Lu Y, Li X. SRY-Box Containing Gene 4 Promotes Liver Steatosis by Upregulation of SREBP-1c. Diabetes 2018; 67:2227-2238. [PMID: 30181160 DOI: 10.2337/db18-0184] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 08/27/2018] [Indexed: 11/13/2022]
Abstract
Obesity is usually associated with an increased risk of nonalcoholic fatty liver disease that is characterized by accumulation of excessive triglyceride (TG) in hepatocytes. However, the factors involved in the obesity-induced hepatosteatosis are poorly defined. Here, we report that SRY-box containing gene 4 (Sox4), a transcription factor that regulates cell proliferation and differentiation, plays an important role in hepatic TG metabolism. Sox4 expression levels are markedly upregulated in livers of obese rodents and humans. Adenovirus-medicated overexpression of Sox4 in the livers of lean mice promotes liver steatosis, whereas liver-specific knockdown of Sox4 ameliorates TG accumulation and improves insulin resistance in obese mice. At the molecular level, we show that Sox4 could directly control the transcription of SREBP-1c gene through binding to its proximal promoter region. Thus, we have identified Sox4 as an important component of hepatic TG metabolism.
Collapse
Affiliation(s)
- Yang Jiao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiejie Zhao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhijian Zhang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Li
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xi Yu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanying Yang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Liu
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, Hubei, China
| | - Shengjie Liao
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, Hubei, China
| | - Duanzhuo Li
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, Hubei, China
| | - Yuxing Wang
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, Hubei, China
| | - Die Zhang
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, Hubei, China
| | - Yulu Chen
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, Hubei, China
| | - Guojun Shi
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
| | - Bin Liu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, Hubei, China
| | - Yan Lu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoying Li
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, and Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Mehdi AM, Hamilton-Williams EE, Cristino A, Ziegler A, Bonifacio E, Le Cao KA, Harris M, Thomas R. A peripheral blood transcriptomic signature predicts autoantibody development in infants at risk of type 1 diabetes. JCI Insight 2018. [PMID: 29515040 DOI: 10.1172/jci.insight.98212] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Autoimmune-mediated destruction of pancreatic islet β cells results in type 1 diabetes (T1D). Serum islet autoantibodies usually develop in genetically susceptible individuals in early childhood before T1D onset, with multiple islet autoantibodies predicting diabetes development. However, most at-risk children remain islet-antibody negative, and no test currently identifies those likely to seroconvert. We sought a genomic signature predicting seroconversion risk by integrating longitudinal peripheral blood gene expression profiles collected in high-risk children included in the BABYDIET and DIPP cohorts, of whom 50 seroconverted. Subjects were followed for 10 years to determine time of seroconversion. Any cohort effect and the time of seroconversion were corrected to uncover genes differentially expressed (DE) in seroconverting children. Gene expression signatures associated with seroconversion were evident during the first year of life, with 67 DE genes identified in seroconverting children relative to those remaining antibody negative. These genes contribute to T cell-, DC-, and B cell-related immune responses. Near-birth expression of ADCY9, PTCH1, MEX3B, IL15RA, ZNF714, TENM1, and PLEKHA5, along with HLA risk score predicted seroconversion (AUC 0.85). The ubiquitin-proteasome pathway linked DE genes and T1D susceptibility genes. Therefore, a gene expression signature in infancy predicts risk of seroconversion. Ubiquitination may play a mechanistic role in diabetes progression.
Collapse
Affiliation(s)
- Ahmed M Mehdi
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Emma E Hamilton-Williams
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Alexandre Cristino
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Anette Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum Munchen, Neuherberg, and Forschergruppe Diabetes, Klinikum rechts der Isar, Institut für Diabetesforschung, Neuherberg, Germany
| | - Ezio Bonifacio
- CRTD-DFG Research Center for Regenerative Therapies Dresden, Medical Faculty Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kim-Anh Le Cao
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Mark Harris
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Australia.,Lady Cilento Children's Hospital, South Brisbane, Australia
| | - Ranjeny Thomas
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, Australia
| |
Collapse
|
8
|
Association of CDKAL1 nucleotide variants with the risk of non-syndromic cleft lip with or without cleft palate. J Hum Genet 2018; 63:397-406. [DOI: 10.1038/s10038-017-0397-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/01/2017] [Accepted: 11/12/2017] [Indexed: 12/24/2022]
|
9
|
Babinsky VN, Hannan FM, Ramracheya RD, Zhang Q, Nesbit MA, Hugill A, Bentley L, Hough TA, Joynson E, Stewart M, Aggarwal A, Prinz-Wohlgenannt M, Gorvin CM, Kallay E, Wells S, Cox RD, Richards D, Rorsman P, Thakker RV. Mutant Mice With Calcium-Sensing Receptor Activation Have Hyperglycemia That Is Rectified by Calcilytic Therapy. Endocrinology 2017; 158:2486-2502. [PMID: 28575322 PMCID: PMC5551547 DOI: 10.1210/en.2017-00111] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 05/30/2017] [Indexed: 12/12/2022]
Abstract
The calcium-sensing receptor (CaSR) is a family C G-protein-coupled receptor that plays a pivotal role in extracellular calcium homeostasis. The CaSR is also highly expressed in pancreatic islet α- and β-cells that secrete glucagon and insulin, respectively. To determine whether the CaSR may influence systemic glucose homeostasis, we characterized a mouse model with a germline gain-of-function CaSR mutation, Leu723Gln, referred to as Nuclear flecks (Nuf). Heterozygous- (CasrNuf/+) and homozygous-affected (CasrNuf/Nuf) mice were shown to have hypocalcemia in association with impaired glucose tolerance and insulin secretion. Oral administration of a CaSR antagonist compound, known as a calcilytic, rectified the glucose intolerance and hypoinsulinemia of CasrNuf/+ mice and ameliorated glucose intolerance in CasrNuf/Nuf mice. Ex vivo studies showed CasrNuf/+ and CasrNuf/Nuf mice to have reduced pancreatic islet mass and β-cell proliferation. Electrophysiological analysis of isolated CasrNuf/Nuf islets showed CaSR activation to increase the basal electrical activity of β-cells independently of effects on the activity of the adenosine triphosphate (ATP)-sensitive K+ (KATP) channel. CasrNuf/Nuf mice also had impaired glucose-mediated suppression of glucagon secretion, which was associated with increased numbers of α-cells and a higher α-cell proliferation rate. Moreover, CasrNuf/Nuf islet electrophysiology demonstrated an impairment of α-cell membrane depolarization in association with attenuated α-cell basal KATP channel activity. These studies indicate that the CaSR activation impairs glucose tolerance by a combination of α- and β-cell defects and also influences pancreatic islet mass. Moreover, our findings highlight a potential application of targeted CaSR compounds for modulating glucose metabolism.
Collapse
Affiliation(s)
- Valerie N. Babinsky
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, United Kingdom
| | - Fadil M. Hannan
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, United Kingdom
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L7 8TX, United Kingdom
| | - Reshma D. Ramracheya
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, United Kingdom
| | - Quan Zhang
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, United Kingdom
| | - M. Andrew Nesbit
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, United Kingdom
- Biomedical Sciences Research Institute, Ulster University, Coleraine BT52 1SA, United Kingdom
| | - Alison Hugill
- Medical Research Council Mammalian Genetics Unit and Mary Lyon Centre, Medical Research Council Harwell Institute, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, United Kingdom
| | - Liz Bentley
- Medical Research Council Mammalian Genetics Unit and Mary Lyon Centre, Medical Research Council Harwell Institute, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, United Kingdom
| | - Tertius A. Hough
- Medical Research Council Mammalian Genetics Unit and Mary Lyon Centre, Medical Research Council Harwell Institute, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, United Kingdom
| | - Elizabeth Joynson
- Medical Research Council Mammalian Genetics Unit and Mary Lyon Centre, Medical Research Council Harwell Institute, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, United Kingdom
| | - Michelle Stewart
- Medical Research Council Mammalian Genetics Unit and Mary Lyon Centre, Medical Research Council Harwell Institute, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, United Kingdom
| | - Abhishek Aggarwal
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna A-1090, Austria
| | | | - Caroline M. Gorvin
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, United Kingdom
| | - Enikö Kallay
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna A-1090, Austria
| | - Sara Wells
- Medical Research Council Mammalian Genetics Unit and Mary Lyon Centre, Medical Research Council Harwell Institute, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, United Kingdom
| | - Roger D. Cox
- Medical Research Council Mammalian Genetics Unit and Mary Lyon Centre, Medical Research Council Harwell Institute, Harwell Science and Innovation Campus, Oxfordshire OX11 0RD, United Kingdom
| | - Duncan Richards
- GlaxoSmithKline Clinical Unit, Cambridge CB2 0GG, United Kingdom
| | - Patrik Rorsman
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, United Kingdom
| | - Rajesh V. Thakker
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LE, United Kingdom
| |
Collapse
|
10
|
Xu EE, Sasaki S, Speckmann T, Nian C, Lynn FC. SOX4 Allows Facultative β-Cell Proliferation Through Repression of Cdkn1a. Diabetes 2017; 66:2213-2219. [PMID: 28495880 DOI: 10.2337/db16-1074] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 05/03/2017] [Indexed: 11/13/2022]
Abstract
The high-mobility group box transcription factor SOX4 is the most highly expressed SOX family protein in pancreatic islets, and mutations in Sox4 are associated with an increased risk of developing type 2 diabetes. We used an inducible β-cell knockout mouse model to test the hypothesis that Sox4 is essential for the maintenance of β-cell number during the development of type 2 diabetes. Knockout of Sox4 at 6 weeks of age resulted in time-dependent worsening of glucose tolerance, impairment of insulin secretion, and diabetes by 30 weeks of age. Immunostaining revealed a decrease in β-cell mass in knockout mice that was caused by a 39% reduction in β-cell proliferation. Gene expression studies revealed that induction of the cell cycle inhibitor Cdkn1a was responsible for the decreased proliferation in the knockout animals. Altogether, this study demonstrates that SOX4 is necessary for adult β-cell replication through direct regulation of the β-cell cycle.
Collapse
Affiliation(s)
- Eric E Xu
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Cell and Developmental Biology Graduate Program, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shugo Sasaki
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Thilo Speckmann
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Cell and Developmental Biology Graduate Program, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cuilan Nian
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Francis C Lynn
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Cell and Developmental Biology Graduate Program, University of British Columbia, Vancouver, British Columbia, Canada
- Departments of Surgery and Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
11
|
Mao L, Wang H, Ma F, Guo Z, He H, Zhou H, Wang N. Exposure to static magnetic fields increases insulin secretion in rat INS-1 cells by activating the transcription of the insulin gene and up-regulating the expression of vesicle-secreted proteins. Int J Radiat Biol 2017; 93:831-840. [PMID: 28593826 DOI: 10.1080/09553002.2017.1332439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE To evaluate the effect of static magnetic fields (SMFs) on insulin secretion and explore the mechanisms underlying exposure to SMF-induced insulin secretion in rat insulinoma INS-1 cells. MATERIALS AND METHODS INS-1 cells were exposed to a 400 mT SMF for 72 h, and the proliferation of INS-1 cells was detected by (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The secretion of insulin was measured with an enzyme linked immunosorbent assays (ELISA), the expression of genes was detected by real-time PCR, and the expression of proteins was measured by Western blotting. RESULTS Exposure to an SMF increased the expression and secretion of insulin by INS-1 cells but did not affect cell proliferation. Moreover, SMF exposure up-regulated the expression of several pancreas-specific transcriptional factors. Specifically, the activity of the rat insulin promoter was enhanced in INS-1 cells exposed to an SMF, and the expression levels of synaptosomal-associated protein 25 (SNAP-25) and syntaxin-1A were up-regulated after exposure to an SMF. CONCLUSIONS SMF exposure can promote insulin secretion in rat INS-1 cells by activating the transcription of the insulin gene and up-regulating the expression of vesicle-secreted proteins.
Collapse
Affiliation(s)
- Libin Mao
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| | - Huiqin Wang
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| | - Fenghui Ma
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| | - Zhixia Guo
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| | - Hongpeng He
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| | - Hao Zhou
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| | - Nan Wang
- a Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology , Tianjin University of Science and Technology , Tianjin , P.R. China
| |
Collapse
|
12
|
Esapa CT, Piret SE, Nesbit MA, Loh NY, Thomas G, Croucher PI, Brown MA, Brown SDM, Cox RD, Thakker RV. Mice with an N-Ethyl-N-Nitrosourea (ENU) Induced Tyr209Asn Mutation in Natriuretic Peptide Receptor 3 (NPR3) Provide a Model for Kyphosis Associated with Activation of the MAPK Signaling Pathway. PLoS One 2016; 11:e0167916. [PMID: 27959934 PMCID: PMC5154531 DOI: 10.1371/journal.pone.0167916] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 11/22/2016] [Indexed: 12/17/2022] Open
Abstract
Non-syndromic kyphosis is a common disorder that is associated with significant morbidity and has a strong genetic involvement; however, the causative genes remain to be identified, as such studies are hampered by genetic heterogeneity, small families and various modes of inheritance. To overcome these limitations, we investigated 12 week old progeny of mice treated with the chemical mutagen N-ethyl-N-nitrosourea (ENU) using phenotypic assessments including dysmorphology, radiography, and dual-energy X-ray absorptiometry. This identified a mouse with autosomal recessive kyphosis (KYLB). KYLB mice, when compared to unaffected littermates, had: thoraco-lumbar kyphosis, larger vertebrae, and increased body length and increased bone area. In addition, female KYLB mice had increases in bone mineral content and plasma alkaline phosphatase activity. Recombination mapping localized the Kylb locus to a 5.5Mb region on chromosome 15A1, which contained 51 genes, including the natriuretic peptide receptor 3 (Npr3) gene. DNA sequence analysis of Npr3 identified a missense mutation, Tyr209Asn, which introduced an N-linked glycosylation consensus sequence. Expression of wild-type NPR3 and the KYLB-associated Tyr209Asn NPR3 mutant in COS-7 cells demonstrated the mutant to be associated with abnormal N-linked glycosylation and retention in the endoplasmic reticulum that resulted in its absence from the plasma membrane. NPR3 is a decoy receptor for C-type natriuretic peptide (CNP), which also binds to NPR2 and stimulates mitogen-activated protein kinase (MAPK) signaling, thereby increasing the number and size of hypertrophic chondrocytes. Histomorphometric analysis of KYLB vertebrae and tibiae showed delayed endochondral ossification and expansion of the hypertrophic zones of the growth plates, and immunohistochemistry revealed increased p38 MAPK phosphorylation throughout the growth plates of KYLB vertebrae. Thus, we established a model of kyphosis due to a novel NPR3 mutation, in which loss of plasma membrane NPR3 expression results in increased MAPK pathway activation, causing elongation of the vertebrae and resulting in kyphosis.
Collapse
Affiliation(s)
- Christopher T. Esapa
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- MRC Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell, Harwell Science and Innovation Campus, Harwell, United Kingdom
| | - Sian E. Piret
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - M. Andrew Nesbit
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nellie Y. Loh
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Gethin Thomas
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | | | - Matthew A. Brown
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Steve D. M. Brown
- MRC Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell, Harwell Science and Innovation Campus, Harwell, United Kingdom
| | - Roger D. Cox
- MRC Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell, Harwell Science and Innovation Campus, Harwell, United Kingdom
| | - Rajesh V. Thakker
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
13
|
Yin C. Molecular mechanisms of Sox transcription factors during the development of liver, bile duct, and pancreas. Semin Cell Dev Biol 2016; 63:68-78. [PMID: 27552918 DOI: 10.1016/j.semcdb.2016.08.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/13/2016] [Accepted: 08/18/2016] [Indexed: 12/15/2022]
Abstract
The liver and pancreas are the prime digestive and metabolic organs in the body. After emerging from the neighboring domains of the foregut endoderm, they turn on distinct differentiation and morphogenesis programs that are regulated by hierarchies of transcription factors. Members of SOX family of transcription factors are expressed in the liver and pancreas throughout development and act upstream of other organ-specific transcription factors. They play key roles in maintaining stem cells and progenitors. They are also master regulators of cell fate determination and tissue morphogenesis. In this review, we summarize the current understanding of SOX transcription factors in mediating liver and pancreas development. We discuss their contribution to adult organ function, homeostasis and injury responses. We also speculate how the knowledge of SOX transcription factors can be applied to improve therapies for liver diseases and diabetes.
Collapse
Affiliation(s)
- Chunyue Yin
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
14
|
Affiliation(s)
- Sebastian Barg
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Alenka Guček
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
15
|
Collins SC, Do HW, Hastoy B, Hugill A, Adam J, Chibalina MV, Galvanovskis J, Godazgar M, Lee S, Goldsworthy M, Salehi A, Tarasov AI, Rosengren AH, Cox R, Rorsman P. Increased Expression of the Diabetes Gene SOX4 Reduces Insulin Secretion by Impaired Fusion Pore Expansion. Diabetes 2016; 65:1952-61. [PMID: 26993066 PMCID: PMC4996324 DOI: 10.2337/db15-1489] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 03/08/2016] [Indexed: 12/27/2022]
Abstract
The transcription factor Sox4 has been proposed to underlie the increased type 2 diabetes risk linked to an intronic single nucleotide polymorphism in CDKAL1 In a mouse model expressing a mutant form of Sox4, glucose-induced insulin secretion is reduced by 40% despite normal intracellular Ca(2+) signaling and depolarization-evoked exocytosis. This paradox is explained by a fourfold increase in kiss-and-run exocytosis (as determined by single-granule exocytosis measurements) in which the fusion pore connecting the granule lumen to the exterior expands to a diameter of only 2 nm, which does not allow the exit of insulin. Microarray analysis indicated that this correlated with an increased expression of the exocytosis-regulating protein Stxbp6. In a large collection of human islet preparations (n = 63), STXBP6 expression and glucose-induced insulin secretion correlated positively and negatively with SOX4 expression, respectively. Overexpression of SOX4 in the human insulin-secreting cell EndoC-βH2 interfered with granule emptying and inhibited hormone release, the latter effect reversed by silencing STXBP6 These data suggest that increased SOX4 expression inhibits insulin secretion and increased diabetes risk by the upregulation of STXBP6 and an increase in kiss-and-run exocytosis at the expense of full fusion. We propose that pharmacological interventions promoting fusion pore expansion may be effective in diabetes therapy.
Collapse
Affiliation(s)
- Stephan C Collins
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, Oxford, U.K. Université de Bourgogne Franche-Comté, Burgundy, France
| | - Hyun Woong Do
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, Oxford, U.K
| | - Benoit Hastoy
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, Oxford, U.K
| | - Alison Hugill
- Mammalian Genetics Unit, MRC Harwell, Oxfordshire, U.K
| | - Julie Adam
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, Oxford, U.K
| | - Margarita V Chibalina
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, Oxford, U.K
| | - Juris Galvanovskis
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, Oxford, U.K. Henry Wellcome Centre for Gene Function, Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, U.K
| | - Mahdieh Godazgar
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, Oxford, U.K
| | - Sheena Lee
- Henry Wellcome Centre for Gene Function, Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, U.K
| | | | - Albert Salehi
- Lund University Diabetes Centre, Department of Clinical Sciences, Skåne University Hospital Malmö, Lund University, Malmö, Sweden Department of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden
| | - Andrei I Tarasov
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, Oxford, U.K. Oxford National Institute of Health Research, Biomedical Research Centre, Churchill Hospital, Oxford, U.K
| | - Anders H Rosengren
- Lund University Diabetes Centre, Department of Clinical Sciences, Skåne University Hospital Malmö, Lund University, Malmö, Sweden
| | - Roger Cox
- Mammalian Genetics Unit, MRC Harwell, Oxfordshire, U.K
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology & Metabolism, Radcliffe Department of Medicine, Oxford, U.K. Department of Neuroscience and Physiology, University of Göteborg, Göteborg, Sweden Oxford National Institute of Health Research, Biomedical Research Centre, Churchill Hospital, Oxford, U.K
| |
Collapse
|
16
|
Goldsworthy M, Bai Y, Li CM, Ge H, Lamas E, Hilton H, Esapa CT, Baker D, Baron W, Juan T, Véniant MM, Lloyd DJ, Cox RD. Haploinsufficiency of the Insulin Receptor in the Presence of a Splice-Site Mutation in Ppp2r2a Results in a Novel Digenic Mouse Model of Type 2 Diabetes. Diabetes 2016; 65:1434-46. [PMID: 26868295 PMCID: PMC5947768 DOI: 10.2337/db15-1276] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 02/07/2016] [Indexed: 12/16/2022]
Abstract
Insulin resistance in mice typically does not manifest as diabetes due to multiple compensatory mechanisms. Here, we present a novel digenic model of type 2 diabetes in mice heterozygous for a null allele of the insulin receptor and an N-ethyl-N-nitrosourea-induced alternative splice mutation in the regulatory protein phosphatase 2A (PP2A) subunit PPP2R2A. Inheritance of either allele independently results in insulin resistance but not overt diabetes. Doubly heterozygous mice exhibit progressive hyperglycemia, hyperinsulinemia, and impaired glucose tolerance from 12 weeks of age without significant increase in body weight. Alternative splicing of Ppp2r2a decreased PPP2R2A protein levels. This reduction in PPP2R2A containing PP2A phosphatase holoenzyme was associated with decreased serine/threonine protein kinase AKT protein levels. Ultimately, reduced insulin-stimulated phosphorylated AKT levels were observed, a result that was confirmed in Hepa1-6, C2C12, and differentiated 3T3-L1 cells knocked down using Ppp2r2a small interfering RNAs. Altered AKT signaling and expression of gluconeogenic genes in the fed state contributed to an insulin resistance and hyperglycemia phenotype. This model demonstrates how genetic changes with individually small phenotypic effects interact to cause diabetes and how differences in expression of hypomorphic alleles of PPP2R2A and potentially other regulatory proteins have deleterious effects and may therefore be relevant in determining diabetes risk.
Collapse
Affiliation(s)
| | - Ying Bai
- Diabetes Group, Medical Research Council Harwell, Oxfordshire, U.K
| | - Chi-Ming Li
- Genome Analysis Unit, Amgen Inc., Thousand Oaks, CA
| | - Huanying Ge
- Genome Analysis Unit, Amgen Inc., Thousand Oaks, CA
| | - Edwin Lamas
- Genome Analysis Unit, Amgen Inc., Thousand Oaks, CA
| | - Helen Hilton
- Protein Core Facility, Medical Research Council Harwell, Oxfordshire, U.K
| | | | - Dan Baker
- Genome Analysis Unit, Amgen Inc., Thousand Oaks, CA
| | - Will Baron
- Genome Analysis Unit, Amgen Inc., Thousand Oaks, CA
| | - Todd Juan
- Genome Analysis Unit, Amgen Inc., Thousand Oaks, CA
| | | | - David J Lloyd
- Department of Metabolic Disorders, Amgen Inc., Thousand Oaks, CA
| | - Roger D Cox
- Diabetes Group, Medical Research Council Harwell, Oxfordshire, U.K.
| |
Collapse
|
17
|
Grapin-Botton A, Seymour PA, Gradwohl G. Pairing-up SOX to kick-start beta cell genesis. Diabetologia 2015; 58:859-61. [PMID: 25733380 DOI: 10.1007/s00125-015-3539-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 02/06/2015] [Indexed: 11/30/2022]
Abstract
The transcription factor SOX9 is regarded as a crucial player in pancreas development, both maintaining progenitors and later being required for beta cell differentiation. However, very little is known about the possible involvement of other SOX family members in such processes. In this issue, the work of Xu et al (DOI: 10.1007/s00125-015-3507-x ) shines a spotlight on SOX4, revealing this factor to be a major player in the beta cell program. Using conditional inactivation in mice, they show that SOX4 shares some functions in progenitors with SOX9, but also plays a distinct role at a later stage of development, during the maturation of endocrine cells. This information is timely as this final maturation process is currently the most challenging to reproduce in vitro when coaxing pluripotent stem cells to convert into beta cells.
Collapse
Affiliation(s)
- Anne Grapin-Botton
- DanStem, University of Copenhagen, 3B Blegdamsvej, DK-2200, Copenhagen N, Denmark,
| | | | | |
Collapse
|
18
|
Xu EE, Krentz NAJ, Tan S, Chow SZ, Tang M, Nian C, Lynn FC. SOX4 cooperates with neurogenin 3 to regulate endocrine pancreas formation in mouse models. Diabetologia 2015; 58:1013-23. [PMID: 25652387 DOI: 10.1007/s00125-015-3507-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 01/09/2015] [Indexed: 10/24/2022]
Abstract
AIMS/HYPOTHESIS The sex-determining region Y (SRY)-related high mobility group (HMG) box (SOX) family of transcription factors is essential for normal organismal development. Despite the longstanding knowledge that many SOX family members are expressed during pancreas development, a role for many of these factors in the establishment of insulin-producing beta cell fate remains to be determined. The aim of this study is to elucidate the role of SOX4 during beta cell development. METHODS We used pancreas and endocrine progenitor mouse knockouts of Sox4 to uncover the roles of SOX4 during pancreas development. Lineage tracing and in vitro models were used to determine how SOX4 regulates beta cell formation and understand the fate of Sox4-null endocrine lineage cells. RESULTS This study demonstrates a progenitor cell-autonomous role for SOX4 in regulating the genesis of beta cells and shows that it is required at multiple stages of the process. SOX4 deletion in the multipotent pancreatic progenitors resulted in impaired endocrine progenitor cell differentiation. Deletion of SOX4 later in the Neurog3-expressing cells also caused reductions in beta cells. Lineage studies showed loss of Sox4 in endocrine progenitors resulted in a block in terminal islet cell differentiation that was attributed to reduction in the production of key beta cell specification factors. CONCLUSIONS/INTERPRETATION These results demonstrate that SOX4 is essential for normal endocrine pancreas development both concomitant with, and downstream of, the endocrine fate decision. In conclusion, these studies position Sox4 temporally in the endocrine differentiation programme and provide a new target for improving in vitro differentiation of glucose-responsive pancreatic beta cells.
Collapse
Affiliation(s)
- Eric E Xu
- Diabetes Research Program, Child and Family Research Institute, A4-184, 950 West 28 Ave, Vancouver, BC, V5Z 4H4, Canada
| | | | | | | | | | | | | |
Collapse
|
19
|
Can the ‘neuron theory’ be complemented by a universal mechanism for generic neuronal differentiation. Cell Tissue Res 2014; 359:343-84. [DOI: 10.1007/s00441-014-2049-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 10/23/2014] [Indexed: 12/19/2022]
|
20
|
Thompson JM, Di Gregorio A. Insulin-like genes in ascidians: findings in Ciona and hypotheses on the evolutionary origins of the pancreas. Genesis 2014; 53:82-104. [PMID: 25378051 DOI: 10.1002/dvg.22832] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 10/13/2014] [Accepted: 10/16/2014] [Indexed: 12/22/2022]
Abstract
Insulin plays an extensively characterized role in the control of sugar metabolism, growth and homeostasis in a wide range of organisms. In vertebrate chordates, insulin is mainly produced by the beta cells of the endocrine pancreas, while in non-chordate animals insulin-producing cells are mainly found in the nervous system and/or scattered along the digestive tract. However, recent studies have indicated the notochord, the defining feature of the chordate phylum, as an additional site of expression of insulin-like peptides. Here we show that two of the three insulin-like genes identified in Ciona intestinalis, an invertebrate chordate with a dual life cycle, are first expressed in the developing notochord during embryogenesis and transition to distinct areas of the adult digestive tract after metamorphosis. In addition, we present data suggesting that the transcription factor Ciona Brachyury is involved in the control of notochord expression of at least one of these genes, Ciona insulin-like 2. Finally, we review the information currently available on insulin-producing cells in ascidians and on pancreas-related transcription factors that might control their expression.
Collapse
Affiliation(s)
- Jordan M Thompson
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, New York, New York
| | | |
Collapse
|
21
|
Jonatan D, Spence JR, Method AM, Kofron M, Sinagoga K, Haataja L, Arvan P, Deutsch GH, Wells JM. Sox17 regulates insulin secretion in the normal and pathologic mouse β cell. PLoS One 2014; 9:e104675. [PMID: 25144761 PMCID: PMC4140688 DOI: 10.1371/journal.pone.0104675] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 07/16/2014] [Indexed: 02/06/2023] Open
Abstract
SOX17 is a key transcriptional regulator that can act by regulating other transcription factors including HNF1β and FOXA2, which are known to regulate postnatal β cell function. Given this, we investigated the role of SOX17 in the developing and postnatal pancreas and found a novel role for SOX17 in regulating insulin secretion. Deletion of the Sox17 gene in the pancreas (Sox17-paLOF) had no observable impact on pancreas development. However, Sox17-paLOF mice had higher islet proinsulin protein content, abnormal trafficking of proinsulin, and dilated secretory organelles suggesting that Sox17-paLOF adult mice are prediabetic. Consistant with this, Sox17-paLOF mice were more susceptible to aged-related and high fat diet-induced hyperglycemia and diabetes. Overexpression of Sox17 in mature β cells using Ins2-rtTA driver mice resulted in precocious secretion of proinsulin. Transcriptionally, SOX17 appears to broadly regulate secretory networks since a 24-hour pulse of SOX17 expression resulted in global transcriptional changes in factors that regulate hormone transport and secretion. Lastly, transient SOX17 overexpression was able to reverse the insulin secretory defects observed in MODY4 animals and restored euglycemia. Together, these data demonstrate a critical new role for SOX17 in regulating insulin trafficking and secretion and that modulation of Sox17-regulated pathways might be used therapeutically to improve cell function in the context of diabetes.
Collapse
Affiliation(s)
- Diva Jonatan
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Jason R. Spence
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, United States of America
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States of America
- Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Anna M. Method
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Matthew Kofron
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Katie Sinagoga
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
| | - Leena Haataja
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Peter Arvan
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Gail H. Deutsch
- Seattle Children’s Hospital, Seattle, WA, United States of America
| | - James M. Wells
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States of America
- * E-mail:
| |
Collapse
|
22
|
Abstract
Embryonic stem (ES) cells have been shown to recapitulate normal developmental stages. They are therefore a highly useful tool in the study of developmental biology. Profiling of ES cell-derived cells has yielded important information about the characteristics of differentiated cells, and allowed the identification of novel marker genes and pathways of differentiation. In this review, we focus on recent results from profiling studies of mouse embryos, human islets, and human ES cell-derived differentiated cells from several research groups. Global gene expression data from mouse embryos have been used to identify novel genes or pathways involved in the developmental process, and to search for transcription factors that regulate direct reprogramming. We introduce gene expression databases of human pancreas cells (Beta Cell Gene Atlas, EuroDia database), and summarize profiling studies of islet- or human ES cell-derived pancreatic cells, with a focus on gene expression, microRNAs, epigenetics, and protein expression. Then, we describe our gene expression profile analyses and our search for novel endoderm, or pancreatic, progenitor marker genes. We differentiated mouse ES cells into mesendoderm, definitive endoderm (DE), mesoderm, ectoderm, and Pdx1-expressing pancreatic lineages, and performed DNA microarray analyses. Genes specifically expressed in DE, and/or in Pdx1-expressing cells, were extracted and their expression patterns in normal embryonic development were studied by in situ hybridization. Out of 54 genes examined, 27 were expressed in the DE of E8.5 mouse embryos, and 15 genes were expressed in distinct domains in the pancreatic buds of E14.5 mouse embryos. Akr1c19, Aebp2, Pbxip1, and Creb3l1 were all novel, and none has been described as being expressed, either in the DE, or in the pancreas. By introducing the profiling results of ES cell-derived cells, the benefits of using ES cells to study early embryonic development will be discussed.
Collapse
Affiliation(s)
- Nobuaki Shiraki
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Kumamoto 860-0811, Japan
| | - Soichiro Ogaki
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Kumamoto 860-0811, Japan
| | - Shoen Kume
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Kumamoto 860-0811, Japan
| |
Collapse
|
23
|
Seelan RS, Mukhopadhyay P, Warner DR, Webb CL, Pisano M, Greene RM. Epigenetic regulation of Sox4 during palate development. Epigenomics 2013; 5:131-46. [PMID: 23566091 DOI: 10.2217/epi.13.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Identification of genes that contribute to secondary palate development provide a better understanding of the etiology of palatal clefts. Gene-expression profiling of the murine palate from gestational days 12-14 (GD12-14), a critical period in palate development, identified Sox4 as a differentially expressed gene. In this study, we have examined if the differential expression of Sox4 in the palate is due to changes in DNA methylation. MATERIALS & METHODS In situ hybridization analysis was used to localize the expression of Sox4 in the developing murine secondary palate. CpG methylation profiling of a 1.8-kb upstream region of Sox4 in the secondary palate from GD12-14 and transfection analysis in murine embryonic maxillary mesenchymal cells using Sox4 deletion, mutant and in vitro methylated plasmid constructs were used to identify critical CpG residues regulating Sox4 expression in the palate. RESULTS Spatiotemporal analysis revealed that Sox4 is expressed in the medial edge epithelium and presumptive rugae-forming regions of the palate from GD12 to GD13. Following palatal shelf fusion on GD14, Sox4 was expressed exclusively in the epithelia of the palatal rugae, structures that serve as signaling centers for the anteroposterior extension of the palate, and that are thought to serve as neural stem cell niches. Methylation of a 1.8-kb region upstream of Sox4, containing the putative promoter, completely eliminated promoter activity. CpG methylation profiling of the 1.8-kb region identified a CpG-poor region (DMR4) that exhibited significant differential methylation during palate development, consistent with changes in Sox4 mRNA expression. Changes in the methylation of DMR4 were attributed primarily to CpGs 83 and 85. CONCLUSION Our studies indicate that Sox4 is an epigenetically regulated gene that likely integrates multiple signaling systems for mediating palatal fusion, palatal extension and/or the maintenance of the neural stem cell niche in the rugae.
Collapse
Affiliation(s)
- Ratnam S Seelan
- University of Louisville, Birth Defects Center, Department of Molecular, Cellular & Craniofacial Biology, ULSD, 501 S. Preston St., Suite 350, Louisville, KY 40202, USA
| | | | | | | | | | | |
Collapse
|
24
|
Jafarnejad SM, Ardekani GS, Ghaffari M, Li G. Pleiotropic function of SRY-related HMG box transcription factor 4 in regulation of tumorigenesis. Cell Mol Life Sci 2013; 70:2677-96. [PMID: 23080209 PMCID: PMC11113534 DOI: 10.1007/s00018-012-1187-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2012] [Revised: 09/10/2012] [Accepted: 10/02/2012] [Indexed: 02/06/2023]
Abstract
In addition to their critical roles in embryonic development, cell fate decision, and differentiation, members of Sox (Sry-related high-mobility group box) family of transcription factors including Sox4 have been implicated in various cancers. Multiple studies have revealed an increased expression along with specific oncogenic function of Sox4 in tumors, while others observed a reduced expression of Sox4 in different types of malignancies and suppression of tumor initiation or progression by this protein. More interestingly, the prognostic value of Sox4 is debated due to obvious differences between various reports as well as inconsistencies within specific studies. This review summarizes our current understanding of Sox4 expression pattern and its transcription-dependent, as well as transcription-independent, functions in tumor initiation or progression and its correlation with patient survival. We also discuss the existing discrepancies between different reports and their possible explanations.
Collapse
Affiliation(s)
- Seyed Mehdi Jafarnejad
- Department of Dermatology and Skin Science, Jack Bell Research Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Gholamreza Safaee Ardekani
- Department of Dermatology and Skin Science, Jack Bell Research Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| | - Mazyar Ghaffari
- The Vancouver Prostate Centre, Vancouver General Hospital, University of British Columbia, Vancouver, BC Canada
| | - Gang Li
- Department of Dermatology and Skin Science, Jack Bell Research Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6 Canada
| |
Collapse
|
25
|
Goldsworthy M, Absalom NL, Schröter D, Matthews HC, Bogani D, Moir L, Long A, Church C, Hugill A, Anstee QM, Goldin R, Thursz M, Hollfelder F, Cox RD. Mutations in Mll2, an H3K4 methyltransferase, result in insulin resistance and impaired glucose tolerance in mice. PLoS One 2013; 8:e61870. [PMID: 23826075 PMCID: PMC3691224 DOI: 10.1371/journal.pone.0061870] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 03/18/2013] [Indexed: 02/03/2023] Open
Abstract
We employed a random mutagenesis approach to identify novel monogenic determinants of type 2 diabetes. Here we show that haplo-insufficiency of the histone methyltransferase myeloid-lineage leukemia (Mll2/Wbp7) gene causes type 2 diabetes in the mouse. We have shown that mice heterozygous for two separate mutations in the SET domain of Mll2 or heterozygous Mll2 knockout mice were hyperglycaemic, hyperinsulinaemic and developed non-alcoholic fatty liver disease. Consistent with previous Mll2 knockout studies, mice homozygous for either ENU mutation (or compound heterozygotes) died during embryonic development at 9.5–14.5 days post coitum. Heterozygous deletion of Mll2 induced in the adult mouse results in a normal phenotype suggesting that changes in chromatin methylation during development result in the adult phenotype. Mll2 has been shown to regulate a small subset of genes, a number of which Neurod1, Enpp1, Slc27a2, and Plcxd1 are downregulated in adult mutant mice. Our results demonstrate that histone H3K4 methyltransferase Mll2 is a component of the genetic regulation necessary for glucose homeostasis, resulting in a specific disease pattern linking chromatin modification with causes and progression of type 2 diabetes, providing a basis for its further understanding at the molecular level.
Collapse
Affiliation(s)
- Michelle Goldsworthy
- Medical Research Council (MRC) Harwell, Diabetes Group, Harwell Science and Innovation Campus, Oxfordshire, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
The role of SRY-related HMG box transcription factor 4 (SOX4) in tumorigenesis and metastasis: friend or foe? Oncogene 2012; 32:3397-409. [PMID: 23246969 DOI: 10.1038/onc.2012.506] [Citation(s) in RCA: 161] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 09/18/2012] [Accepted: 09/18/2012] [Indexed: 12/29/2022]
Abstract
Development and progression of cancer are mediated by alterations in transcriptional networks, resulting in a disturbed balance between the activity of oncogenes and tumor suppressor genes. Transcription factors have the capacity to regulate global transcriptional profiles, and are consequently often found to be deregulated in their expression and function during tumorigenesis. Sex-determining region Y-related high-mobility-group box transcription factor 4 (SOX4) is a member of the group C subfamily of the SOX transcription factors and has a critical role during embryogenesis, where its expression is widespread and controls the development of numerous tissues. SOX4 expression is elevated in a wide variety of tumors, including leukemia, colorectal cancer, lung cancer and breast cancer, suggesting a fundamental role in the development of these malignancies. In many cancers, deregulated expression of this developmental factor has been correlated with increased cancer cell proliferation, cell survival, inhibition of apoptosis and tumor progression through the induction of an epithelial-to-mesenchymal transition and metastasis. However, in a limited subset of tumors, SOX4 has also been reported to act as a tumor suppressor. These opposing roles suggest that the outcome of SOX4 activation depends on the cellular context and the tumor origin. Indeed, SOX4 expression, transcriptional activity and target gene specificity can be controlled by signaling pathways, including the transforming growth factor-β and the WNT pathway, as well as at the post-translational level through regulation of protein stability and interaction with specific cofactors, such as TCF, syntenin-1 and p53. Here, we provide an overview of our current knowledge concerning the role of SOX4 in tumor development and progression.
Collapse
|
27
|
Becker TS, Rinkwitz S. Zebrafish as a genomics model for human neurological and polygenic disorders. Dev Neurobiol 2012; 72:415-28. [PMID: 21465670 DOI: 10.1002/dneu.20888] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Whole exome sequencing and, to a lesser extent, genome-wide association studies, have provided unprecedented advances in identifying genes and candidate genomic regions involved in the development of human disease. Further progress will come from sequencing the entire genome of multiple patients and normal controls to evaluate overall mutational burden and disease risk. A major challenge will be the interpretation of the resulting data and distinguishing true pathogenic mutations from rare benign variants.While in model organisms such as the zebrafish,mutants are sought that disrupt the function of individual genes, human mutations that cause, or are associated with, the development of disease, are often not acting in a Mendelian fashion, are frequently of small effect size, are late onset, and may reside in noncoding parts of the genome. The zebrafish model is uniquely poised for understanding human coding- and noncoding variants because of its sequenced genome, a large body of knowledge on gene expression and function, rapid generation time, and easy access to embryos. A critical advantage is the ease of zebrafish transgenesis, both for the testing of human regulatory DNA driving expression of fluorescent reporter proteins, and the expression of mutated disease-associated human proteins in specific neurons to rapidly model aspects of neurological disorders. The zebrafish affords progress both through its model genome and it is rapidly developing transparent model vertebrate embryo.
Collapse
Affiliation(s)
- Thomas S Becker
- Sydney Medical School, University of Sydney, Camperdown, Australia.
| | | |
Collapse
|
28
|
Brown FC, Scott N, Rank G, Collinge JE, Vadolas J, Vickaryous N, Whitelaw N, Whitelaw E, Kile BT, Jane SM, Curtis DJ. ENU mutagenesis identifies the first mouse mutants reproducing human β-thalassemia at the genomic level. Blood Cells Mol Dis 2012; 50:86-92. [PMID: 23040355 DOI: 10.1016/j.bcmd.2012.09.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 09/11/2012] [Indexed: 12/24/2022]
Abstract
Forward genetic screens have been performed in many species to identify phenotypes in specific organ systems. We have undertaken a large-scale N-ethyl-N-nitrosourea (ENU) mutagenesis screen to identify dominant mutations that perturb erythropoiesis in mice. Mutant mice that displayed an erythrocyte mean cell volume (MCV) greater than three standard deviations from the population mean were identified. Two of these lines, RBC13 and RBC14, displayed a hypochromic, microcytic anemia, accompanied by a marked reticulocytosis, splenomegaly and diminished red cell survival. Timed pregnancies from heterozygous intercrosses revealed that a quarter of the embryos displayed severe anemia and did not survive beyond embryonic day (E) 18.5, consistent with homozygous β-thalassemia. Genetic complementation studies with a β-thalassemia mouse line reproduced the embryonic lethality in compound heterozygotes and a genomic custom capture array and massively parallel sequencing of the β-globin locus identified the causative mutations. The RBC13 line displayed a nonsense mutation at codon 40 in exon 2 of the β-major gene, invoking parallels with the common β(0)39 thalassemia mutation seen in humans. The RBC14 line exhibited a mutation at the polyadenylation signal of the β-major gene, exactly replicating a human β-thalassemia mutation. The RBC13 and RBC14 lines are the first β-thalassemia mouse models that reproduce human β-thalassemia at the genomic level, and as such highlight the power of ENU mutagenesis screens in generating mouse models of human disease.
Collapse
Affiliation(s)
- Fiona C Brown
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC 3004, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
ENU mutagenesis is a forward genetics strategy in which random mutagenesis and phenotypic screening is used to identify genes based on the phenotype induced when they are mutated. A modifier screen is a type of screen in which mice with a pre-existing phenotype are utilized to identify mutations that can enhance or suppress this phenotype. This approach has the potential to uncover missing pathway members, reveal novel genetic interactions, and pinpoint new drug targets. Considerations when planning a suppressor screen include current knowledge, genomic footprint, penetrance, variance, robustness, latency of the starting phenotype, viability, fertility, genetic background and ENU tolerance of starting strain, screening assay, mouse numbers required, and mutation identification strategy. Practical advice on each of these is provided in this review. Curr. Protoc. Mouse Biol. 2:75-87 © 2012 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Marina R Carpinelli
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Australia.,Hearing Co-Operative Research Centre, Melbourne, Australia.,Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Rachel A Burt
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Australia.,Hearing Co-Operative Research Centre, Melbourne, Australia.,Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Benjamin T Kile
- Cancer and Hematology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Australia
| |
Collapse
|
30
|
Esapa CT, Hough TA, Testori S, Head RA, Crane EA, Chan CPS, Evans H, Bassett JHD, Tylzanowski P, McNally EG, Carr AJ, Boyde A, Howell PGT, Clark A, Williams GR, Brown MA, Croucher PI, Nesbit MA, Brown SDM, Cox RD, Cheeseman MT, Thakker RV. A mouse model for spondyloepiphyseal dysplasia congenita with secondary osteoarthritis due to a Col2a1 mutation. J Bone Miner Res 2012; 27:413-28. [PMID: 22028304 DOI: 10.1002/jbmr.547] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Progeny of mice treated with the mutagen N-ethyl-N-nitrosourea (ENU) revealed a mouse, designated Longpockets (Lpk), with short humeri, abnormal vertebrae, and disorganized growth plates, features consistent with spondyloepiphyseal dysplasia congenita (SEDC). The Lpk phenotype was inherited as an autosomal dominant trait. Lpk/+ mice were viable and fertile and Lpk/Lpk mice died perinatally. Lpk was mapped to chromosome 15 and mutational analysis of likely candidates from the interval revealed a Col2a1 missense Ser1386Pro mutation. Transient transfection of wild-type and Ser1386Pro mutant Col2a1 c-Myc constructs in COS-7 cells and CH8 chondrocytes demonstrated abnormal processing and endoplasmic reticulum retention of the mutant protein. Histology revealed growth plate disorganization in 14-day-old Lpk/+ mice and embryonic cartilage from Lpk/+ and Lpk/Lpk mice had reduced safranin-O and type-II collagen staining in the extracellular matrix. The wild-type and Lpk/+ embryos had vertical columns of proliferating chondrocytes, whereas those in Lpk/Lpk mice were perpendicular to the direction of bone growth. Electron microscopy of cartilage from 18.5 dpc wild-type, Lpk/+, and Lpk/Lpk embryos revealed fewer and less elaborate collagen fibrils in the mutants, with enlarged vacuoles in the endoplasmic reticulum that contained amorphous inclusions. Micro-computed tomography (CT) scans of 12-week-old Lpk/+ mice revealed them to have decreased bone mineral density, and total bone volume, with erosions and osteophytes at the joints. Thus, an ENU mouse model with a Ser1386Pro mutation of the Col2a1 C-propeptide domain that results in abnormal collagen processing and phenotypic features consistent with SEDC and secondary osteoarthritis has been established.
Collapse
Affiliation(s)
- Christopher T Esapa
- Academic Endocrine Unit, Nuffield Department of Clinical Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Headington, Oxford, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Mouse models and type 2 diabetes: translational opportunities. Mamm Genome 2011; 22:390-400. [PMID: 21713584 DOI: 10.1007/s00335-011-9345-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 06/07/2011] [Indexed: 01/14/2023]
Abstract
Type 2 diabetes prevalence is increasing worldwide. Treatments are available, but glycaemic control is not always effective in many patients. Better models are needed to create new and improved therapies and to expand our understanding of how type 2 diabetes begins and progresses. Translational research involves the transformation of knowledge from basic scientific discoveries to impacting on public health. This can allow identification of novel molecular mechanisms underlying the disease which can lead to preventative measures, biomarkers for diagnosis, or future therapies. Generation of genetically modified mice has allowed us to investigate the function of genes and develop reproducible models in which the phenotype of the animal can be tested. Mouse models have already given us insight into glucose metabolism and insulin secretion, identified novel pathways, and have been used to confirm genome-wide association studies. In this review we discuss the use of the mouse to clarify human genome-wide association study loci, understand genes and pathways involved in type 2 diabetes, and uncover novel targets for drug discovery.
Collapse
|
32
|
Boogerd CJJ, Wong LYE, van den Boogaard M, Bakker ML, Tessadori F, Bakkers J, 't Hoen PAC, Moorman AF, Christoffels VM, Barnett P. Sox4 mediates Tbx3 transcriptional regulation of the gap junction protein Cx43. Cell Mol Life Sci 2011; 68:3949-61. [PMID: 21538160 PMCID: PMC3214269 DOI: 10.1007/s00018-011-0693-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 12/01/2010] [Accepted: 04/14/2011] [Indexed: 11/28/2022]
Abstract
Tbx3, a T-box transcription factor, regulates key steps in development of the heart and other organ systems. Here, we identify Sox4 as an interacting partner of Tbx3. Pull-down and nuclear retention assays verify this interaction and in situ hybridization reveals Tbx3 and Sox4 to co-localize extensively in the embryo including the atrioventricular and outflow tract cushion mesenchyme and a small area of interventricular myocardium. Tbx3, SOX4, and SOX2 ChIP data, identify a region in intron 1 of Gja1 bound by all tree proteins and subsequent ChIP experiments verify that this sequence is bound, in vivo, in the developing heart. In a luciferase reporter assay, this element displays a synergistic antagonistic response to co-transfection of Tbx3 and Sox4 and in vivo, in zebrafish, drives expression of a reporter in the heart, confirming its function as a cardiac enhancer. Mechanistically, we postulate that Sox4 is a mediator of Tbx3 transcriptional activity.
Collapse
Affiliation(s)
- C J J Boogerd
- Heart Failure Research Centre, Academic Medical Centre, University of Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kim HD, Choe HK, Chung S, Kim M, Seong JY, Son GH, Kim K. Class-C SOX transcription factors control GnRH gene expression via the intronic transcriptional enhancer. Mol Endocrinol 2011; 25:1184-96. [PMID: 21527504 DOI: 10.1210/me.2010-0332] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
GnRH is a pivotal hypothalamic neurohormone governing reproduction and sexual development. Because transcriptional regulation is crucial for the spatial and temporal expression of the GnRH gene, a region approximately 3.0 kb upstream of the mammalian GnRH promoter has been extensive studied. In the present study, we demonstrate a transcription-enhancer located in the first intron (intron A) region of the GnRH gene. This transcriptional enhancer harbors putative sex-determining region Y-related high-mobility-group box (SOX) family transcription factor-binding sites, which are well conserved across many mammalian species. The class-C SOX member proteins (SOX-C) (SOX4 and SOX11) specifically augment this transcriptional activation by binding to these SOX-binding sites. In accordance, SOX11 is highly enriched in immortalized GnRH-producing GT1-1 cells, and suppression of its expression significantly decreases GnRH gene expression as well as GnRH secretion. Chromatin immunoprecipitation shows that endogenous SOX-C factors recognize and bind to the intronic enhancer in GT1-1 cells and the hypothalamus. Accompanying immunohistochemical analysis demonstrates that SOX4 or SOX11 are highly expressed in the majority of hypothalamic GnRH neurons in adult mice. Taken together, these findings demonstrate that SOX-C transcription factors function as important transcriptional regulators of cell type-specific GnRH gene expression by acting on the intronic transcriptional enhancer.
Collapse
Affiliation(s)
- Hee-Dae Kim
- Department of Biological Sciences, Seoul National University, Brain Research Center for the 21st Century Frontier Program in Neuroscience, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
34
|
Ogaki S, Harada S, Shiraki N, Kume K, Kume S. An expression profile analysis of ES cell-derived definitive endodermal cells and Pdx1-expressing cells. BMC DEVELOPMENTAL BIOLOGY 2011; 11:13. [PMID: 21362171 PMCID: PMC3058101 DOI: 10.1186/1471-213x-11-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Accepted: 03/01/2011] [Indexed: 12/16/2022]
Abstract
Background We developed an efficient in vitro method to differentiate mouse ES cells into the definitive endoderm (DE) and then Pdx1-expressing pancreatic lineages using mesodermal-derived supporting cells, M15. Using this method, resulting ES cell-derived DE and Pdx1-expressing cells were isolated by cell sorting, and their gene expression profiles were investigated with DNA microarray. Genes that were specifically expressed in DE and/or in Pdx1-expressing cells were extracted and their expression patterns in normal embryonic development were studied. Results Genes whose expression increased in DE and Pdx1 positive cells compared to the undifferentiated ES cells were chosen and in situ hybridizations were performed. Out of 54 genes examined, 27 were expressed in the DE of E8.5 mouse embryos and 15 genes were expressed in distinct domains in the pancreatic buds of E14.5 embryos. Among those genes expressed were Foxq1, CpM, Foxp4, Pcdh1, and Zmiz1, which were previously reported in other endodermal tissues. Genes, such as Parm1, Tmem184a, Hipk2 and Sox4 were reported to be expressed during early pancreatic development. Nptx2, C2cd4b, Tcf7l2 and Kiss1r were reported to be associated with beta cell or pancreatic functions in the adult. Akr1c19, Aebp2, Pbxip1 and Creb3l1, were novel and have not been described as being expressed either in DE or the pancreas. Conclusions We identified 27 genes, including 4 novel genes expressed in DE and pancreatic progenitor cells during normal development using an ES cell in vitro differentiation system. These results showed that DE cells and Pdx1/GFP-expressing cells obtained from our M15 based differentiation method mimic cells during the normal developmental processes. Additionally, ES cells are an excellent model for studies of early developmental processes.
Collapse
Affiliation(s)
- Soichiro Ogaki
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Kumamoto, Japan
| | | | | | | | | |
Collapse
|
35
|
Abstract
The use of mouse models in medical research has greatly contributed to our understanding of the development of type 2 diabetes mellitus and the mechanisms of disease progression in the context of insulin resistance and β-cell dysfunction. Maintenance of glucose homeostasis involves a complex interplay of many genes and their actions in response to exogenous stimuli. In recent years, the availability of large population-based cohorts and the capacity to genotype enormous numbers of common genetic variants have driven various large-scale genome-wide association studies, which has greatly accelerated the identification of novel genes likely to be involved in the development of type 2 diabetes. The increasing demand for verifying novel genes is met by the timely development of new mouse resources established as various collaborative projects involving major transgenic and phenotyping centres and laboratories worldwide. The surge of new data will ultimately enable translational research into potential improvement and refinement of current type 2 diabetes therapy options, and hopefully restore quality of life for patients.
Collapse
|
36
|
Nguyen N, Judd LM, Kalantzis A, Whittle B, Giraud AS, van Driel IR. Random mutagenesis of the mouse genome: a strategy for discovering gene function and the molecular basis of disease. Am J Physiol Gastrointest Liver Physiol 2011; 300:G1-11. [PMID: 20947703 PMCID: PMC3774088 DOI: 10.1152/ajpgi.00343.2010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mutagenesis of mice with N-ethyl-N-nitrosourea (ENU) is a phenotype-driven approach to unravel gene function and discover new biological pathways. Phenotype-driven approaches have the advantage of making no assumptions about the function of genes and their products and have been successfully applied to the discovery of novel gene-phenotype relationships in many physiological systems. ENU mutagenesis of mice is used in many large-scale and more focused projects to generate and identify novel mouse models for the study of gene functions and human disease. This review examines the strategies and tools used in ENU mutagenesis screens to efficiently generate and identify functional mutations.
Collapse
Affiliation(s)
- Nhung Nguyen
- 1Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne;
| | - Louise M. Judd
- 2Gastrointestinal Research in Inflammation and Pathology Laboratory, Murdoch Children's Research Institute, Melbourne; and
| | - Anastasia Kalantzis
- 2Gastrointestinal Research in Inflammation and Pathology Laboratory, Murdoch Children's Research Institute, Melbourne; and
| | - Belinda Whittle
- 3Australian Phenomics Facility, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Andrew S. Giraud
- 2Gastrointestinal Research in Inflammation and Pathology Laboratory, Murdoch Children's Research Institute, Melbourne; and
| | - Ian R. van Driel
- 1Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne;
| |
Collapse
|
37
|
Yu H, Huang J, Qiao N, Green CD, Han JDJ. Evaluating diabetes and hypertension disease causality using mouse phenotypes. BMC SYSTEMS BIOLOGY 2010; 4:97. [PMID: 20642857 PMCID: PMC2917432 DOI: 10.1186/1752-0509-4-97] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2010] [Accepted: 07/20/2010] [Indexed: 01/11/2023]
Abstract
Background Genome-wide association studies (GWAS) have found hundreds of single nucleotide polymorphisms (SNPs) associated with common diseases. However, it is largely unknown what genes linked with the SNPs actually implicate disease causality. A definitive proof for disease causality can be demonstration of disease-like phenotypes through genetic perturbation of the genes or alleles, which is obviously a daunting task for complex diseases where only mammalian models can be used. Results Here we tapped the rich resource of mouse phenotype data and developed a method to quantify the probability that a gene perturbation causes the phenotypes of a disease. Using type II diabetes (T2D) and hypertension (HT) as study cases, we found that the genes, when perturbed, having high probability to cause T2D and HT phenotypes tend to be hubs in the interactome networks and are enriched for signaling pathways regulating metabolism but not metabolic pathways, even though the genes in these metabolic pathways are often the most significantly changed in expression levels in these diseases. Conclusions Compared to human genetic disease-based predictions, our mouse phenotype based predictors greatly increased the coverage while keeping a similarly high specificity. The disease phenotype probabilities given by our approach can be used to evaluate the likelihood of disease causality of disease-associated genes and genes surrounding disease-associated SNPs.
Collapse
Affiliation(s)
- Hong Yu
- Chinese Academy of Sciences Key Laboratory of Molecular Developmental Biology, Center for Molecular Systems Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Lincui East Road, Beijing 100101, China
| | | | | | | | | |
Collapse
|
38
|
Long-range gene regulation links genomic type 2 diabetes and obesity risk regions to HHEX, SOX4, and IRX3. Proc Natl Acad Sci U S A 2009; 107:775-80. [PMID: 20080751 DOI: 10.1073/pnas.0911591107] [Citation(s) in RCA: 169] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Genome-wide association studies identified noncoding SNPs associated with type 2 diabetes and obesity in linkage disequilibrium (LD) blocks encompassing HHEX-IDE and introns of CDKAL1 and FTO [Sladek R, et al. (2007) Nature 445:881-885; Steinthorsdottir V, et al. (2007) Nat. Genet 39:770-775; Frayling TM, et al. (2007) Science 316:889-894]. We show that these LD blocks contain highly conserved noncoding elements and overlap with the genomic regulatory blocks of the transcription factor genes HHEX, SOX4, and IRX3. We report that human highly conserved noncoding elements in LD with the risk SNPs drive expression in endoderm or pancreas in transgenic mice and zebrafish. Both HHEX and SOX4 have recently been implicated in pancreas development and the regulation of insulin secretion, but IRX3 had no prior association with pancreatic function or development. Knockdown of its orthologue in zebrafish, irx3a, increased the number of pancreatic ghrelin-producing epsilon cells and decreased the number of insulin-producing beta-cells and glucagon-producing alpha-cells, thereby suggesting a direct link of pancreatic IRX3 function to both obesity and type 2 diabetes.
Collapse
|
39
|
McDonald E, Krishnamurthy M, Goodyer CG, Wang R. The Emerging Role of SOX Transcription Factors in Pancreatic Endocrine Cell Development and Function. Stem Cells Dev 2009; 18:1379-88. [DOI: 10.1089/scd.2009.0240] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Affiliation(s)
- Erin McDonald
- Children’s Health Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Mansa Krishnamurthy
- Children’s Health Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Cynthia G. Goodyer
- Department of Pediatrics, McGill University Health Centre, Children’s Hospital Research Institute, Montreal, Quebec, Canada
| | - Rennian Wang
- Children’s Health Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
40
|
Insulin secretion from beta-cells is affected by deletion of nicotinamide nucleotide transhydrogenase. Methods Enzymol 2009; 457:451-80. [PMID: 19426883 DOI: 10.1016/s0076-6879(09)05025-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
Nicotinamide nucleotide transhydrogenase (NNT) is an inner mitochondrial membrane transmembrane protein involved in regenerating NADPH, coupled with proton translocation across the inner membrane. We have shown that a defect in Nnt function in the mouse, and specifically within the beta-cell, leads to a reduction in insulin secretion. This chapter describes methods for examining Nnt function in the mouse. This includes generating in vivo models with point mutations and expression of Nnt by transgenesis, and making in vitro models, by silencing of gene expression. In addition, techniques are described to measure insulin secretion, calcium and hydrogen peroxide concentrations, membrane potential, and NNT activity. These approaches and techniques can also be applied to other genes of interest.
Collapse
|
41
|
Current literature in diabetes. Diabetes Metab Res Rev 2009; 25:i-x. [PMID: 19219862 DOI: 10.1002/dmrr.918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
42
|
Abstract
The major forms of diabetes are characterized by pancreatic islet beta-cell dysfunction and decreased beta-cell numbers, raising hope for cell replacement therapy. Although human islet transplantation is a cell-based therapy under clinical investigation for the treatment of type 1 diabetes, the limited availability of human cadaveric islets for transplantation will preclude its widespread therapeutic application. The result has been an intense focus on the development of alternate sources of beta cells, such as through the guided differentiation of stem or precursor cell populations or the transdifferentiation of more plentiful mature cell populations. Realizing the potential for cell-based therapies, however, requires a thorough understanding of pancreas development and beta-cell formation. Pancreas development is coordinated by a complex interplay of signaling pathways and transcription factors that determine early pancreatic specification as well as the later differentiation of exocrine and endocrine lineages. This review describes the current knowledge of these factors as they relate specifically to the emergence of endocrine beta cells from pancreatic endoderm. Current therapeutic efforts to generate insulin-producing beta-like cells from embryonic stem cells have already capitalized on recent advances in our understanding of the embryonic signals and transcription factors that dictate lineage specification and will most certainly be further enhanced by a continuing emphasis on the identification of novel factors and regulatory relationships.
Collapse
Affiliation(s)
- Jennifer M. Oliver-Krasinski
- Institute for Diabetes, Obesity and Metabolism and the Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Doris A. Stoffers
- Institute for Diabetes, Obesity and Metabolism and the Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|