1
|
Mitchell SB, Aydemir TB. ZIP Family Zinc Transporters: Emerging Players in Pancreatic β Cell Function and Insulin Regulation. J Nutr 2025:S0022-3166(25)00363-3. [PMID: 40516652 DOI: 10.1016/j.tjnut.2025.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/20/2025] [Accepted: 06/10/2025] [Indexed: 06/16/2025] Open
Abstract
Zinc is an essential micronutrient with diverse catalytic, structural, and regulatory roles across various life forms. Its essentiality for human health was recognized in the 1960s, but advancements in understanding the functions of zinc at the tissue, cell, and subcellular levels have accelerated, particularly with the identification of zinc transporters. Zinc homeostasis is primarily facilitated by two families of transporters, the SLC30A/ZNT (zinc transporters) and SLC39A/ZIP (Zrt-Irt-Like Proteins). Among these, the ZNT family transporter ZNT8 has been well-studied for its involvement in insulin production, secretion, and the viability of pancreatic β cells. However, the roles of ZIP family transporters in β cell insulin-related functions remain less explored. There have been studies implicating regulatory roles of ZIP4, ZIP5, ZIP6, and ZIP7 in β cells and emerging evidence for the involvement of ZIP8 and ZIP14 in β cell function. Despite these insights, the limited number of studies on ZIP family transporters highlights the need to consolidate existing literature to identify gaps and establish targeted, comprehensive research approaches that can further elucidate their critical roles in cellular zinc homeostasis and insulin metabolism. In this review, we first address the role of zinc in insulin production, secretion, and action. Second, we discuss the known ZIP transporters that potentially facilitate zinc delivery to specific cell compartments, focusing on literature addressing zinc and zinc transporters specifically relevant to insulin and glucose metabolism.
Collapse
|
2
|
Fabiola León-Galván M, Medina-Rojas DS. DPP-IV and FAS inhibitory peptides: therapeutic alternative against diabesity. J Diabetes Metab Disord 2025; 24:100. [PMID: 40224529 PMCID: PMC11985882 DOI: 10.1007/s40200-025-01613-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/21/2025] [Indexed: 04/15/2025]
Abstract
Diabesity is a modern epidemic that indicates a strong association between obesity and diabetes. Key enzymes have been identified in the development and progression of both diseases, DPP-IV in glucose uptake and FAS in fatty acid synthesis. In both cases, the molecular mechanisms of how each one acts separately have been described, and which are the key inhibitory drugs and molecules for each one. However, although it is known that there is an association between both clinically and molecularly, the mechanism has not been elucidated; therefore, this review focuses on proposing a mechanism of convergence of DPP-IV and FAS in diabesity, and the possible mode of action in which bioactive peptides obtained from plant and animal sources can inhibit these two enzymes in a similar way as drugs do.
Collapse
Affiliation(s)
- Ma. Fabiola León-Galván
- Food Department, Proteomics and Gene Expression Laboratory, University of Guanajuato, Life Science Division, Campus Irapuato-Salamanca, Ex Hacienda el Copal, Carretera Irapuato-Silao km 9.0, Irapuato, C.P 36500 Guanajuato México
- Graduate Program in Biosciences, Proteomics and Gene Expression Laboratory, University of Guanajuato, Life Science Division, Campus Irapuato-Salamanca, Ex Hacienda el Copal, Carretera Irapuato-Silao km 9.0, Irapuato, C.P 36500 Guanajuato México
| | - Daniela Sarahi Medina-Rojas
- Graduate Program in Biosciences, Proteomics and Gene Expression Laboratory, University of Guanajuato, Life Science Division, Campus Irapuato-Salamanca, Ex Hacienda el Copal, Carretera Irapuato-Silao km 9.0, Irapuato, C.P 36500 Guanajuato México
| |
Collapse
|
3
|
Bineid MM, Ventura EF, Samidoust A, Radha V, Anjana RM, Sudha V, Walton GE, Mohan V, Vimaleswaran KS. A Systematic Review of the Effect of Gene-Lifestyle Interactions on Metabolic-Disease-Related Traits in South Asian Populations. Nutr Rev 2025; 83:1061-1082. [PMID: 39283705 PMCID: PMC12066952 DOI: 10.1093/nutrit/nuae115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025] Open
Abstract
CONTEXT Recent data from the South Asian subregion have raised concern about the dramatic increase in the prevalence of metabolic diseases, which are influenced by genetic and lifestyle factors. OBJECTIVE The aim of this systematic review was to summarize the contemporary evidence for the effect of gene-lifestyle interactions on metabolic outcomes in this population. DATA SOURCES PubMed, Web of Science, and SCOPUS databases were searched up until March 2023 for observational and intervention studies investigating the interaction between genetic variants and lifestyle factors such as diet and physical activity on obesity and type 2 diabetes traits. DATA EXTRACTION Of the 14 783 publications extracted, 15 were deemed eligible for inclusion in this study. Data extraction was carried out independently by 3 investigators. The quality of the included studies was assessed using the Appraisal Tool for Cross-Sectional Studies (AXIS), the Risk Of Bias In Non-randomized Studies-of Interventions (ROBINS-I), and the methodological quality score for nutrigenetics studies. DATA ANALYSIS Using a narrative synthesis approach, the findings were presented in textual and tabular format. Together, studies from India (n = 8), Pakistan (n = 3), Sri Lanka (n = 1), and the South Asian diaspora in Singapore and Canada (n = 3) reported 543 gene-lifestyle interactions, of which 132 (∼24%) were statistically significant. These results were related to the effects of the interaction of genetic factors with physical inactivity, poor sleep habits, smoking, and dietary intake of carbohydrates, protein, and fat on the risk of metabolic disease in this population. CONCLUSIONS The findings of this systematic review provide evidence of gene-lifestyle interactions impacting metabolic traits within the South Asian population. However, the lack of replication and correction for multiple testing and the small sample size of the included studies may limit the conclusiveness of the evidence. Note, this paper is part of the Nutrition Reviews Special Collection on Precision Nutrition. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration No. CRD42023402408.
Collapse
Affiliation(s)
- Manahil M Bineid
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading RG6 6DZ, United Kingdom
| | - Eduard F Ventura
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), 46980 Valencia, Spain
| | - Aryan Samidoust
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading RG6 6DZ, United Kingdom
| | - Venkatesan Radha
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Centre for Advanced Research on Diabetes, Chennai 603103, India
| | - Ranjit Mohan Anjana
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Centre for Advanced Research on Diabetes, Chennai 603103, India
- Department of Foods, Nutrition and Dietetics Research, Madras Diabetes Research Foundation, Chennai 600086, India
- Department of Diabetology, Dr Mohan’s Diabetes Specialties Centre, IDF Centre of Excellence in Diabetes Care, Chennai 600086, India
| | - Vasudevan Sudha
- Department of Foods, Nutrition and Dietetics Research, Madras Diabetes Research Foundation, Chennai 600086, India
| | - Gemma E Walton
- Food Microbial Sciences Unit, Department of Food and Nutritional Sciences, University of Reading, Whiteknights, Reading RG6 6AP, United Kingdom
| | - Viswanathan Mohan
- Department of Molecular Genetics, Madras Diabetes Research Foundation, ICMR Centre for Advanced Research on Diabetes, Chennai 603103, India
- Department of Foods, Nutrition and Dietetics Research, Madras Diabetes Research Foundation, Chennai 600086, India
- Department of Diabetology, Dr Mohan’s Diabetes Specialties Centre, IDF Centre of Excellence in Diabetes Care, Chennai 600086, India
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading RG6 6DZ, United Kingdom
- The Institute for Food, Nutrition and Health (IFNH), University of Reading, Reading RG6 6AH, United Kingdom
| |
Collapse
|
4
|
Lee H, Fernandes M, Lee J, Merino J, Kwak SH. Exploring the shared genetic landscape of diabetes and cardiovascular disease: findings and future implications. Diabetologia 2025; 68:1087-1100. [PMID: 40088285 PMCID: PMC12069157 DOI: 10.1007/s00125-025-06403-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/28/2025] [Indexed: 03/17/2025]
Abstract
Diabetes is a rapidly growing global health concern projected to affect one in eight adults by 2045, which translates to roughly 783 million people. The profound metabolic alterations often present in dysglycaemia significantly increase the risk of cardiovascular complications. While genetic susceptibility plays a crucial role in diabetes and its vascular complications, identifying genes and molecular mechanisms that influence both diseases simultaneously has proven challenging. A key reason for this challenge is the pathophysiological heterogeneity underlying these diseases, with multiple processes contributing to different forms of diabetes and specific cardiovascular complications. This molecular heterogeneity has limited the effectiveness of large-scale genome-wide association studies (GWAS) in identifying shared underlying mechanisms. Additionally, our limited knowledge of the causal genes, cell types and disease-relevant states through which GWAS signals operate has hindered the discovery of common molecular pathways. This review highlights recent advances in genetic epidemiology, including studies of causal associations that have uncovered genetic and molecular factors influencing both dysglycaemia and cardiovascular complications. We explore how disease subtyping approaches can be critical in pinpointing the unique molecular signatures underlying both diabetes and cardiovascular complications. Finally, we address critical research gaps and future opportunities to advance our understanding of both diseases and translate these discoveries into tangible benefits for patient care and population health.
Collapse
Affiliation(s)
- Hyunsuk Lee
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Maria Fernandes
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jeongeun Lee
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Korea
| | - Jordi Merino
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
- Diabetes Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
| | - Soo Heon Kwak
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
5
|
Nikolitsa EK, Kontou PI, Bagos PG. metacp: a versatile software package for combining dependent or independent p-values. BMC Bioinformatics 2025; 26:109. [PMID: 40253343 PMCID: PMC12008841 DOI: 10.1186/s12859-025-06126-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 04/01/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND We present metacp an open-source software package which implements an abundance of statistical methods for the combination of both independent p-values, with methods such as Fisher's, Stouffer's and Edgington's, and dependent p-values, with methods such as Brown's method and the Cauchy Combination Test. RESULTS The tool is available in Python and STATA, it is very fast, and it is easy to use, requiring only minimal input. It offers a useful resource for combining both independent and dependent p-values, responding to diverse analytical needs for practitioners performing meta-analyses and bioinformaticians developing tools for a variety of applications. Depending on the input data it can be used for gene-based testing, for analysis of multiple traits in GWAS, or for combining diverse multi-omics data such as those of a TWAS, a colocalization or an RNA-seq study. CONCLUSIONS Compared to other similar packages (like poolr or metap), metacp implements the largest collection of statistical methods for this problem, offering users the flexibility to choose from a wide variety of approaches. Being available both as a standalone Python tool and as a STATA command, metacp is accessible to a broad and diverse audience, including practitioners conducting meta-analyses across various fields and bioinformaticians developing new tools where p-value combination is a crucial component.
Collapse
Affiliation(s)
- Evgenia K Nikolitsa
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 35100, Lamia, Greece
| | | | - Pantelis G Bagos
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 35100, Lamia, Greece.
| |
Collapse
|
6
|
Krawczyk J, O'Connor W, Vendramini P, Schell M, Biddinger KJ, Pengo G, Fougeray T, Aragam KG, Haigis M, Lamers WH, Tsai LT, Biddinger SB. The Diabetes Gene Tcf7l2 Organizes Gene Expression in the Liver and Regulates Amino Acid Metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.647067. [PMID: 40291732 PMCID: PMC12026580 DOI: 10.1101/2025.04.03.647067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
TCF7L2 harbors the strongest genetic association with diabetes identified thus far. However, its function in liver has remained unclear. Here, we find using mice with liver-specific deletion, that Tcf7l2 plays a central role in maintaining hepatic zonation. That is, in the normal liver, many genes show gradients of expression across the liver lobule; in the absence of Tcf7l2 , these gradients collapse. One major consequence is the disorganization of glutamine metabolism, with a loss of the glutamine production program, ectopic expression of the glutamine consumption program, and a decrease in glutamine levels. In parallel, metabolomic profiling shows glutamine to be the most significantly decreased metabolite in individuals harboring the rs7903146 variant in TCF7L2 . Taken together, these data indicate that hepatic TCF7L2 has a secondary role in glycemic control, but a primary role in maintaining transcriptional architecture and glutamine homeostasis.
Collapse
|
7
|
Duarte GBS, Pascoal GDFL, Rogero MM. Polymorphisms Involved in Insulin Resistance and Metabolic Inflammation: Influence of Nutrients and Dietary Interventions. Metabolites 2025; 15:245. [PMID: 40278374 PMCID: PMC12029114 DOI: 10.3390/metabo15040245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/17/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Insulin resistance (IR) is a metabolic disorder characterized by an impaired response to insulin. This condition is associated with excess adiposity and metabolic inflammation, contributing to an increased risk for related chronic diseases. Single-nucleotide polymorphisms (SNPs) can affect genes related to metabolic pathways which are related to IR and the individual response to nutrients and dietary patterns, affecting metabolic inflammation and insulin sensitivity. This narrative review explores the current evidence on interactions between genetic variants and dietary factors, specifically their effects in modulating IR and metabolic inflammation. A comprehensive search of the literature was conducted in PubMed, Google Scholar, and Web of Science, and a total of 95 articles were reviewed. The key findings reveal that SNPs in the TCF7L2, ADIPOQ, and TNF genes significantly influence metabolic responses and modulate the effects of the Mediterranean diet on biomarkers of inflammation and IR. Genotype-dependent variations in IR and inflammation biomarkers were observed in the response to different diets for SNPs in the TCF7L2, ADIPOQ, and TNF genes. Additionally, polygenic risk scores (PRSs) can also predict the response to the intake of nutrients and specific diets, and offer a promising tool for assessing genetic predisposition to IR. This review underscores the pivotal role of an individual's genetic background in the effects of their nutrient intake and in the responses to dietetic interventions, thereby laying the foundation for personalized and effective nutritional strategies tailored to each individual's necessity in mitigating IR and its associated risk factors for chronic diseases.
Collapse
Affiliation(s)
| | | | - Marcelo Macedo Rogero
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo 01246-904, Brazil; (G.B.S.D.); (G.d.F.L.P.)
| |
Collapse
|
8
|
Lai CQ, Gervis JE, Parnell LD, Lichtenstein AH, Ordovas JM. Changes in triglyceride-rich lipoprotein particle profiles in response to one-week on a low fat or Mediterranean diet by TCF7L2 rs7903146 genotype: a randomized crossover dietary intervention trial. GENES & NUTRITION 2025; 20:4. [PMID: 40050721 PMCID: PMC11884055 DOI: 10.1186/s12263-025-00763-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/19/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND The TCF7L2 gene is a significant genetic factor contributing to the risk of metabolic and cardiovascular diseases (CVD). We previously found that subjects with the TT genotype of TCF7L2 rs7903146 variant, who consume a low-fat diet (LF) had a higher incidence of stroke than subjects with the CC genotype. Yet this association was abolished in subjects with the TT genotype who consumed a Mediterranean-type diet (MetD). However, the mechanism by which MetD diet modulates the association between TCF7L2 and CVD risk is unclear. This study aims to validate these findings under real-world conditions and clinical practice to elucidate the biological mechanisms involved in this correlation. METHODS Thirty-five participants with BMI ranging from 27 to 34 kg/m2 were recruited based on rs7903146 genotype. Of those consented to participate, 21 had the CC and 14 had the TT genotype. Participants were randomly assigned to two dietary intervention groups, ensuring an equal distribution of CC and TT carriers. Each participant followed one of two diets (LF or MetD) for one week, followed by a 10-day washout period before switching to the other diet for one week. Blood samples were collected before and after each diet for metabolomic analysis using nuclear magnetic resonance (NMR) spectroscopy. The differential effect of the diets on triglyceride-rich lipoproteins was determined based on TCF7L2 genotype. RESULTS The MetD significantly reduced triglyceride-rich lipoprotein concentrations compared to the LF diet. After consuming the LF diet, TT carriers exhibited more small VLDL particles, potentially contributing to CVD risk compared to CC carriers. However, this difference in risk was not observed with the MetD. Furthermore, the order in which the two diets were crossed affected the triglyceride-rich lipoprotein profile, with LF-MetD regimen showing a stronger effect on triglyceride-rich lipoproteins (TRL) levels than the MetD-LF regimen. CONCLUSIONS Our findings suggest that rs7903146 TT carriers benefit more from a MetD than a LF diet in terms of their triglyceride-rich lipoprotein profile, which may reduce their risk of CVD. These results support the notion that genotype is a factor in determining the extent to which the MetD affects cardiovascular health.
Collapse
Affiliation(s)
- Chao-Qiang Lai
- USDA ARS, Precision Nutrition Directive, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA.
| | - Julie E Gervis
- Diet & Chronic Disease Prevention Directive, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Laurence D Parnell
- USDA ARS, Precision Nutrition Directive, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Alice H Lichtenstein
- Diet & Chronic Disease Prevention Directive, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Jose M Ordovas
- Precision Nutrition Directive, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA.
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain.
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
9
|
Li P, Ye H, Guo F, Zheng J, Shen W, Xie D, Shi S, Zhang Y, Fa Y, Zhao Z. Construction of cynomolgus monkey type 2 diabetes models by combining genetic prediction model with high-energy diet. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167616. [PMID: 39672349 DOI: 10.1016/j.bbadis.2024.167616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2D) is a significant health concern. Research using non-human primates, which develop T2D with similar symptoms and pancreatic changes as humans, is crucial but limited by long timelines and low success rates. RESULTS We targeted capture sequenced 61 normal and 81 T2D cynomolgus monkeys using a primer panel that captured 269 potential regulatory regions potentially associated with T2D in the cynomolgus monkey genome. 80 variants were identified to be associated with T2D and were used to construct a genetic prediction model. Among 8 machine learning algorithms tested, we found that the best prediction performance was achieve when the model using support vector machine with polynomial kernel as the machine learning algorithm (AUC = 0.933). Including age and sex in this model did not significantly improve the prediction performance. Using the genetic prediction model, we further screened 22 monkeys and found 13 were high risk while 9 were low risk. After feeding the 22 monkeys with high-energy food for 32 weeks, we found all the 9 low risk monkeys did not develop T2D while 4 out of 13 high risk monkeys (31 %) develop T2D. CONCLUSIONS This method greatly increased the success rate of establishing T2D monkey models while decreased the time needed compared to traditional methods. Therefore, we developed a new high-efficiency method to establish T2D monkey models by combining the genetic prediction model and high-energy diet, which will greatly contribute to the research on the clinical characteristics, pathogenesis, complications and potential new treatments.
Collapse
Affiliation(s)
- Ping Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Huahu Ye
- Academy of Military Medical Sciences, Beijing, China
| | - Feng Guo
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Jianhua Zheng
- Academy of Military Medical Sciences, Beijing, China
| | - Wenlong Shen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Dejian Xie
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Shu Shi
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Yan Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China.
| | - Yunzhi Fa
- Academy of Military Medical Sciences, Beijing, China
| | - Zhihu Zhao
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
10
|
Chen SY, Tsai RY, Tseng TJ, Chen CC. Elevated PTPN3 expression in type 2 diabetes mellitus: Insights from genetic and experimental analyses. Biomed Rep 2025; 22:53. [PMID: 39926046 PMCID: PMC11803341 DOI: 10.3892/br.2025.1931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/17/2024] [Indexed: 02/11/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with chronic hyperglycemia, leading to severe complications, including increased risk of cancer. Protein tyrosine phosphatase non-receptor type 3 (PTPN3) is implicated in both T2DM and cancer progression. The aim of the present study was to investigate the role of PTPN3 genetic polymorphisms and expression in patients with T2DM, as well as to examine changes in body weight, blood glucose levels, and hepatic PTPN3 expression in db/db obese mice in comparison with control mice at 4, 16 and 32 weeks. A total of 469 patients with T2DM and 1,699 healthy control subjects were analyzed for PTPN3 genetic polymorphisms using blood samples. Additionally, the body weight of genetically diabetic obese db/db mice and genotype control mice, and their fasting blood glucose and PTPN3 mRNA and protein expression levels were assessed in the respective liver tissues at different stages of T2DM progression (4, 16 and 32 weeks) using reverse transcription-quantitative PCR, western blot and immunohistochemistry staining analyses. The allele C frequency of rs75235286 (82.1 vs. 79.1%, P=0.044) and allele G frequency of rs17202063 (82.8% vs. 79.5%, P=0.027) in PTPN3 SNPs differed significantly between T2DM patients and healthy controls. Additionally, the body weight of db/db mice and blood glucose levels were significantly increased from the 4th to 32nd week compared with control mice. Furthermore, db/db mice exhibited significantly elevated hepatic mRNA and protein expression levels of PTPN3 compared with control mice, especially at the 32nd week. Taken together, these findings suggested that an increased level of PTPN3 expression may serve a role in the progression of diabetic complications in patients with T2DM, highlighting the importance of further investigation into PTPN3 as a potential therapeutic target to decrease cancer risk and enhance treatment outcomes in T2DM.
Collapse
Affiliation(s)
- Shih-Yin Chen
- School of Chinese Medicine, China Medical University, Taichung 404328, Taiwan, R.O.C
- Genetics Center, Department of Medical Research, China Medical University Hospital, Taichung 404328, Taiwan, R.O.C
| | - Ru-Yin Tsai
- Department of Anatomy, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan, R.O.C
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung 40201, Taiwan, R.O.C
| | - To-Jung Tseng
- Department of Anatomy, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan, R.O.C
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung 40201, Taiwan, R.O.C
| | - Chin-Chang Chen
- Department of Anatomy, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan, R.O.C
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung 40201, Taiwan, R.O.C
| |
Collapse
|
11
|
Al-Rawaf HA, Gabr SA, Alghadir T, Alghadir F, Iqbal A, Alghadir AH. Correlation between circulating microRNAs and vascular biomarkers in type 2 diabetes based upon physical activity: a biochemical analytic study. BMC Endocr Disord 2025; 25:55. [PMID: 40016689 PMCID: PMC11866858 DOI: 10.1186/s12902-025-01855-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/21/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND This research investigated how physical activity (PA) might impact the expression of several microRNAs, specifically miR-126, miR-146a, miR-34a, miR-124a, miR-155, and miR-221, in the blood of elderly individuals with type 2 diabetes (T2D). Additionally, the study examined the relationship between these microRNAs and markers of vascular endothelial dysfunction, including vascular endothelial growth factor (VEGF), apolipoprotein A-I (apoA-I), and apolipoprotein B (apoB), to assess their potential in the prevention, early detection, and treatment of diabetes. METHODS This correlational observational study involved 100 male participants, aged between 18 and 65 years, all of whom had been living with type 2 diabetes (T2D) for over six years. The participants were divided into three groups: inactive, moderate, and active, depending on their level of physical activity (PA). Real-time PCR and immunoassays were employed to measure the expression of selected miRNAs, as well as VEGF, apoA-I, apoB, and diabetic management indicators. PA levels were determined using ACTi graph GT1M accelerometer (model WAM 7164; Fort Walton Beach, FL) and energy expenditure was measured in the form of metabolic equivalent (MET) by indirect calorimetry method. RESULTS The expression levels of miR-146a, miR-34a, and miR-124a were significantly higher in patients with higher physical activity, while no such increase was observed for the other miRNAs in less active participants. Additionally, PA-active individuals showed a more pronounced decrease in fasting blood sugar (FBS), insulin resistance (IR), fasting insulin (FINS), HOMA-IR, HbA1c (%), and levels of VEGF, apoAI, apoB, and the apoB/apoA-I ratio. The alteration in miRNA expression was positively associated with physical activity, VEGF, apoAI, apoB, the apoB/apoA-I ratio, and diabetes-related metrics, while being inversely related to BMI. CONCLUSIONS In diabetic patients with higher physical activity levels, circulating miR-146a, miR-34a, and miR-124a showed elevated expression, accompanied by a notable decrease in vascular biomarkers, including apoAI, apoB, and the apoB/apoA-I ratio. The findings revealed a strong correlation between these vascular biomarkers and the physiological responses of miR-146a, miR-34a, and miR-124a, though larger studies are required to validate these results further. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Hadeel A Al-Rawaf
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
- Rehabilitation Research Chair, Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| | - Sami A Gabr
- Rehabilitation Research Chair, Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| | - Talal Alghadir
- College of Medicine, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Faisal Alghadir
- College of Medicine, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Amir Iqbal
- Rehabilitation Research Chair, Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia.
| | - Ahmad H Alghadir
- Rehabilitation Research Chair, Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| |
Collapse
|
12
|
Martin SS, Aday AW, Allen NB, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Bansal N, Beaton AZ, Commodore-Mensah Y, Currie ME, Elkind MSV, Fan W, Generoso G, Gibbs BB, Heard DG, Hiremath S, Johansen MC, Kazi DS, Ko D, Leppert MH, Magnani JW, Michos ED, Mussolino ME, Parikh NI, Perman SM, Rezk-Hanna M, Roth GA, Shah NS, Springer MV, St-Onge MP, Thacker EL, Urbut SM, Van Spall HGC, Voeks JH, Whelton SP, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2025 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2025; 151:e41-e660. [PMID: 39866113 DOI: 10.1161/cir.0000000000001303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2025 AHA Statistical Update is the product of a full year's worth of effort in 2024 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. This year's edition includes a continued focus on health equity across several key domains and enhanced global data that reflect improved methods and incorporation of ≈3000 new data sources since last year's Statistical Update. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
13
|
Le Collen L, Froguel P, Bonnefond A. Towards the recognition of oligogenic forms of type 2 diabetes. Trends Endocrinol Metab 2025; 36:109-117. [PMID: 38955653 DOI: 10.1016/j.tem.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 07/04/2024]
Abstract
The demarcation between monogenic and polygenic type 2 diabetes (T2D) is less distinct than previously believed. Notably, recent research has highlighted a new entity, that we suggest calling oligogenic forms of T2D, serving as a genetic link between these two forms. In this opinion article, we have reviewed scientific advances that suggest categorizing genes involved in oligogenic T2D. Research focused on polygenic T2D has faced challenges in deepening our comprehension of the pathophysiology of T2D due to the inability to directly establish causal links between a signal and the molecular mechanisms underlying the disease. However, the study of oligogenic forms of T2D has illuminated distinct causal connections between genes and disease risk, thereby indicating potential new drug targets.
Collapse
Affiliation(s)
- Lauriane Le Collen
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille University Hospital, Lille, France; University of Lille, Lille, France; Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, Nutrition, and Metabolism, University Hospital of Nancy, Nancy, France
| | - Philippe Froguel
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille University Hospital, Lille, France; University of Lille, Lille, France; Department of Metabolism, Imperial College London, Hammersmith Hospital, London, UK
| | - Amélie Bonnefond
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille University Hospital, Lille, France; University of Lille, Lille, France; Department of Metabolism, Imperial College London, Hammersmith Hospital, London, UK.
| |
Collapse
|
14
|
Bonnefond A, Florez JC, Loos RJF, Froguel P. Dissection of type 2 diabetes: a genetic perspective. Lancet Diabetes Endocrinol 2025; 13:149-164. [PMID: 39818223 DOI: 10.1016/s2213-8587(24)00339-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/11/2024] [Accepted: 10/30/2024] [Indexed: 01/18/2025]
Abstract
Diabetes is a leading cause of global mortality and disability, and its economic burden is substantial. This Review focuses on type 2 diabetes, which makes up 90-95% of all diabetes cases. Type 2 diabetes involves a progressive loss of insulin secretion often alongside insulin resistance and metabolic syndrome. Although obesity and a sedentary lifestyle are considerable contributors, research over the last 25 years has shown that type 2 diabetes develops on a predisposing genetic background, with family and twin studies indicating considerable heritability (ie, 31-72%). This Review explores type 2 diabetes from a genetic perspective, highlighting insights into its pathophysiology and the implications for precision medicine. More specifically, the traditional understanding of type 2 diabetes genetics has focused on a dichotomy between monogenic and polygenic forms. However, emerging evidence suggests a continuum that includes monogenic, oligogenic, and polygenic contributions, revealing their complementary roles in type 2 diabetes pathophysiology. Recent genetic studies provide deeper insights into disease mechanisms and pave the way for precision medicine approaches that could transform type 2 diabetes management. Additionally, the effect of environmental factors on type 2 diabetes, particularly from epigenetic modifications, adds another layer of complexity to understanding and addressing this multifaceted disease.
Collapse
Affiliation(s)
- Amélie Bonnefond
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France; Department of Metabolism, Imperial College London, London, UK.
| | - Jose C Florez
- Center for Genomic Medicine and Diabetes Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA; Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Ruth J F Loos
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Philippe Froguel
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France; Department of Metabolism, Imperial College London, London, UK.
| |
Collapse
|
15
|
Sirivarasai J, Tristitworn P, Shantavasinkul PC, Roytrakul S, Chansirikarnjana S, Ruangritchankul S, Chanprasertyothin S, Charernwat P, Panpunuan P, Sura T, Sritara P. Genetic Polymorphism of Zinc Transporter-8 Gene (SLC30A8), Serum Zinc Concentrations, and Proteome Profiles Related to Type 2 Diabetes in Elderly. J Clin Med 2025; 14:790. [PMID: 39941463 PMCID: PMC11818826 DOI: 10.3390/jcm14030790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Background and Aims: Older adults are particularly susceptible to type 2 diabetes mellitus (T2DM) due to factors such as age-related insulin resistance, decreased physical activity, and deficiency of micronutrients, especially zinc. Studies have suggested that the risk allele of the zinc transporter 8 gene (SLC30A8) single-nucleotide poly-morphism (SNP) rs13266634 may contribute to T2DM susceptibility in addition to the complex protein interactions and alterations in the protein expressions and modifications associated with T2DM. This study was implemented to study the associations between SLC30A8 polymorphism, serum zinc levels, and the profiles of proteins differentially expressed in nondiabetic (n = 116) and prediabetic/diabetic (n = 149) subjects. Methods: SNP genotyping using TaqMan® assay and proteomic analysis by LC-MS/MS were performed in each group. Results: The results showed a higher risk of diabetes in individuals with the risk genotype CC accompanied by a low serum zinc level than in those with other genotypes. Profiles of proteins differentially expressed between the groups were identified and shown to be particularly associated with zinc-related functions, zinc transporter 8, and glucose metabolism. Proteins exclusively expressed in prediabetes/diabetes were assigned to a Reactome pathway related to zinc transporter and insulin processing. Conclusions: Our findings suggest that individuals carrying at least one copy of SLC30A8 rs13266634 accompanied by a low serum zinc level might be susceptible to T2DM, which could be due to alterations in insulin signaling and zinc metabolism. Understanding this relationship deepens our understanding of the genetic and molecular mechanisms underlying T2DM risk, offering potential targets for therapeutic intervention and prevention strategies.
Collapse
Affiliation(s)
- Jintana Sirivarasai
- Nutrition Unit, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Pimvaree Tristitworn
- Master of Science Program in Nutrition, Faculty of Medicine Ramathibodi Hospital and Institute of Nutrition, Mahidol University, Bangkok 10400, Thailand;
| | | | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology, 113 Thailand Science Park, Pathum Thani 12120, Thailand;
| | - Sirintorn Chansirikarnjana
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (P.C.S.); (S.C.); (S.R.); (P.C.); (P.P.); (T.S.); (P.S.)
| | - Sirasa Ruangritchankul
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (P.C.S.); (S.C.); (S.R.); (P.C.); (P.P.); (T.S.); (P.S.)
| | - Suwannee Chanprasertyothin
- Research & Innovation, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand;
| | - Piangporn Charernwat
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (P.C.S.); (S.C.); (S.R.); (P.C.); (P.P.); (T.S.); (P.S.)
| | - Pachara Panpunuan
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (P.C.S.); (S.C.); (S.R.); (P.C.); (P.P.); (T.S.); (P.S.)
| | - Thanyachai Sura
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (P.C.S.); (S.C.); (S.R.); (P.C.); (P.P.); (T.S.); (P.S.)
| | - Piyamitr Sritara
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand; (P.C.S.); (S.C.); (S.R.); (P.C.); (P.P.); (T.S.); (P.S.)
| |
Collapse
|
16
|
Mancl JM, Liang WG, Bayhi NL, Wei H, Carragher B, Potter CS, Tang WJ. Characterization and modulation of human insulin degrading enzyme conformational dynamics to control enzyme activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.30.630732. [PMID: 39803525 PMCID: PMC11722313 DOI: 10.1101/2024.12.30.630732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Insulin degrading enzyme (IDE) is a dimeric 110 kDa M16A zinc metalloprotease that degrades amyloidogenic peptides diverse in shape and sequence, including insulin, amylin, and amyloid-β, to prevent toxic amyloid fibril formation. IDE has a hollow catalytic chamber formed by four homologous subdomains organized into two ~55 kDa N- and C- domains (IDE-N and IDE-C, respectively), in which peptides bind, unfold, and are repositioned for proteolysis. IDE is known to transition between a closed state, poised for catalysis, and an open state, able to release cleavage products and bind new substrate. Here, we present five cryoEM structures of the IDE dimer at 3.0-4.1 Å resolution, obtained in the presence of a sub-saturating concentration of insulin. Analysis of the heterogeneity within the particle populations comprising these structures combined with all-atom molecular dynamics (MD) simulations permitted a comprehensive characterization of IDE conformational dynamics. Our analysis identified the structural basis and key residues for these dynamics that were not revealed by IDE static structures. Notably arginine-668 serves as a molecular latch mediating the open-close transition and facilitates key protein motions through charge-swapping interactions at the IDE-N/C interface. Our size-exclusion chromatography-coupled small-angle X-ray scattering and enzymatic assays of an arginine-668 to alanine mutant indicate a profound alteration of conformational dynamics and catalytic activity. Taken together, this work highlights the power of integrating experimental and computational methodologies to understand protein dynamics, offers the molecular basis of unfoldase activity of IDE, and provides a new path forward towards the development of substrate-specific modulators of IDE activity.
Collapse
Affiliation(s)
- Jordan M. Mancl
- Ben-May Institute for Cancer Research, The University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, USA
| | - Wenguang G. Liang
- Ben-May Institute for Cancer Research, The University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, USA
- Simons Electron Microscopy Center, New York Structural Biology Center, 89 Convent Ave, New York, New York, 10027, USA
| | - Nicholas L. Bayhi
- Biophysics Science Graduate Program, The University of Chicago, 929 East 57th street, Chicago, Illinois 60637, USA
| | - Hui Wei
- Simons Electron Microscopy Center, New York Structural Biology Center, 89 Convent Ave, New York, New York, 10027, USA
| | - Bridget Carragher
- Simons Electron Microscopy Center, New York Structural Biology Center, 89 Convent Ave, New York, New York, 10027, USA
- Current affiliation - Chan Zuckerberg Institute for Advanced Biological Imaging
| | - Clinton S. Potter
- Simons Electron Microscopy Center, New York Structural Biology Center, 89 Convent Ave, New York, New York, 10027, USA
- Current affiliation - Chan Zuckerberg Institute for Advanced Biological Imaging
| | - Wei-Jen Tang
- Ben-May Institute for Cancer Research, The University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, USA
| |
Collapse
|
17
|
Jia W, Chan JC, Wong TY, Fisher EB. Diabetes in China: epidemiology, pathophysiology and multi-omics. Nat Metab 2025; 7:16-34. [PMID: 39809974 DOI: 10.1038/s42255-024-01190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 11/25/2024] [Indexed: 01/16/2025]
Abstract
Although diabetes is now a global epidemic, China has the highest number of affected people, presenting profound public health and socioeconomic challenges. In China, rapid ecological and lifestyle shifts have dramatically altered diabetes epidemiology and risk factors. In this Review, we summarize the epidemiological trends and the impact of traditional and emerging risk factors on Chinese diabetes prevalence. We also explore recent genetic, metagenomic and metabolomic studies of diabetes in Chinese, highlighting their role in pathogenesis and clinical management. Although heterogeneity across these multidimensional areas poses major analytic challenges in classifying patterns or features, they have also provided an opportunity to increase the accuracy and specificity of diagnosis for personalized treatment and prevention. National strategies and ongoing research are essential for improving diabetes detection, prevention and control, and for personalizing care to alleviate societal impacts and maintain quality of life.
Collapse
Affiliation(s)
- Weiping Jia
- Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute for Proactive Healthcare, Shanghai Jiao Tong University, Shanghai, China.
| | - Juliana Cn Chan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences and Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Tien Y Wong
- Tsinghua Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
- Singapore National Eye Center, SingHealth, Singapore, Singapore
| | - Edwin B Fisher
- Peers for Progress, Department of Health Behavior, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
18
|
Mueller JW, Thomas P, Dalgaard LT, da Silva Xavier G. Sulfation pathways in the maintenance of functional beta-cell mass and implications for diabetes. Essays Biochem 2024; 68:509-522. [PMID: 39290144 PMCID: PMC11625869 DOI: 10.1042/ebc20240034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/21/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024]
Abstract
Diabetes Type 1 and Type 2 are widely occurring diseases. In spite of a vast amount of biomedical literature about diabetic processes in general, links to certain biological processes are only becoming evident these days. One such area of biology is the sulfation of small molecules, such as steroid hormones or metabolites from the gastrointestinal tract, as well as larger biomolecules, such as proteins and proteoglycans. Thus, modulating the physicochemical propensities of the different sulfate acceptors, resulting in enhanced solubility, expedited circulatory transit, or enhanced macromolecular interaction. This review lists evidence for the involvement of sulfation pathways in the maintenance of functional pancreatic beta-cell mass and the implications for diabetes, grouped into various classes of sulfated biomolecule. Complex heparan sulfates might play a role in the development and maintenance of beta-cells. The sulfolipids sulfatide and sulfo-cholesterol might contribute to beta-cell health. In beta-cells, there are only very few proteins with confirmed sulfation on some tyrosine residues, with the IRS4 molecule being one of them. Sulfated steroid hormones, such as estradiol-sulfate and vitamin-D-sulfate, may facilitate downstream steroid signaling in beta-cells, following de-sulfation. Indoxyl sulfate is a metabolite from the intestine, that causes kidney damage, contributing to diabetic kidney disease. Finally, from a technological perspective, there is heparan sulfate, heparin, and chondroitin sulfate, that all might be involved in next-generation beta-cell transplantation. Sulfation pathways may play a role in pancreatic beta-cells through multiple mechanisms. A more coherent understanding of sulfation pathways in diabetes will facilitate discussion and guide future research.
Collapse
Affiliation(s)
- Jonathan Wolf Mueller
- Department of Metabolism and Systems Science, University of Birmingham, Birmingham, U.K
| | - Patricia Thomas
- Department of Metabolism and Systems Science, University of Birmingham, Birmingham, U.K
| | | | | |
Collapse
|
19
|
Piro A, Luo Y, Zhang Z, Ye W, Kang F, Xie L, Wang Y, Dai FF, Gaisano HY, Rocheleau JV, Prentice KJ, Wheeler MB. Beta cell specific ZnT8 gene deficiency and resulting loss in zinc content significantly improve insulin secretion. Mol Cell Endocrinol 2024; 594:112376. [PMID: 39321953 DOI: 10.1016/j.mce.2024.112376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/06/2024] [Accepted: 09/22/2024] [Indexed: 09/27/2024]
Abstract
Zinc transporter 8 (ZnT8) is highly expressed in pancreatic beta cells, localizes to insulin secretory granules (ISG), and regulates zinc content. ZnT8 gene polymorphisms have revealed a relationship between ZnT8 activity and type 2 diabetes (T2D) risk, however, the role of beta-cell ZnT8 is not well understood. A beta cell specific ZnT8 knockout (ZnT8 BKO) mouse model was investigated. ZnT8 BKO islets showed significantly reduced ZnT8 gene expression and reduced zinc content. Compared to controls, ZnT8 BKO mice displayed significantly elevated plasma insulin levels and improved glucose tolerance following acute insulin resistance induced via S961. Glucose stimulated insulin secretion from isolated ZnT8 BKO pancreatic islets revealed enhanced insulin secretion capacity. The difference in insulin secretion between ZnT8 BKO and control islets was negated upon zinc supplementation, and the inhibitory effect of zinc on insulin secretion was confirmed in human islets. These results indicate that the loss of ZnT8 activity and accompanying reduced cellular zinc are associated with increased insulin secretion capacity. The reduction in secreted insulin content upon zinc supplementation in ZnT8 BKO islets suggests that ISG-released zinc normally tempers insulin secretion.
Collapse
Affiliation(s)
- Anthony Piro
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yihan Luo
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ziyi Zhang
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenyue Ye
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Fei Kang
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Li Xie
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yufeng Wang
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON M5G 1L7, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Feihan F Dai
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Herbert Y Gaisano
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan V Rocheleau
- Advanced Diagnostics, Toronto General Hospital Research Institute, Toronto, ON M5G 1L7, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Kacey J Prentice
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michael B Wheeler
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
20
|
Zeng Q, Liu J, Liu X, Liu N, Wu W, Watson RG, Zou D, Wei Y, Guo R. Association between genetic polymorphisms and gestational diabetes mellitus susceptibility in a Chinese population. Front Endocrinol (Lausanne) 2024; 15:1397423. [PMID: 39659616 PMCID: PMC11628248 DOI: 10.3389/fendo.2024.1397423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Background Although the association between HHEX, IGF2BP2, and FTO polymorphisms and the risk of GDM has been investigated in several studies, the findings have been inconsistent across different populations. The study aimed to investigate the association between genetic polymorphisms and GDM risk in a Chinese population. Methods 502 control volunteers and 500 GDM patients were enrolled. IGF2BP2 rs11705701 and rs4402960, FTO rs9939609, and HHEX rs1111875 and rs5015480 were all genotyped using the SNPscan™ genotyping assay. The independent sample t-test, logistic regression, and chi-square test were used to assess the variations in genotype and allele and their relationships with the risk of GDM. The blood glucose level, gestational week of delivery, and newborn weight were compared using a one-way ANOVA. Results After adjusting for confounding factors, the results show that the rs1111875 heterozygous (OR=1.370; 95% CI: 1.040-1.805; P = 0.025) and overdominant (OR=1.373; 95% CI: 1.049-1.796; P = 0. 021) models are significantly associated with an increased risk of GDM, especially for the age ≥ 30 years group: heterozygote (OR=1.646; 95% CI: 1.118-2.423; P=0.012) and overdominant (OR=1.553; 95% CI: 1.064-2.266; P = 0.022) models. In the age ≥ 30 years, the rs5015480 overdominant model (OR=1.595; 95% CI: 1.034-2.459; P = 0.035) and the rs9939609 heterozygote model (OR=1.609; 95% CI: 1.016-2.550; P=0.043), allele (OR=1. 504; 95% CI: 1.006-2.248; P = 0.047), dominant model (OR=1.604; 95% CI: 1.026-2.505; P = 0.038), and overdominant model (OR=1.593; 95% CI: 1.007-2.520; P = 0.047) were associated with a significantly increased risk of GDM; Additionally, people with the TC genotype of rs1111875 had a substantially higher 1-hour blood glucose level than TT genotype (P < 0.05). The results of the meta-analysis showed that the A allele of rs11705701 was associated with an increased risk of diabetes mellitus (P < 0.05). Conclusion The study indicates that the TC genotype of rs1111875 is linked to a higher risk of GDM, particularly in women aged 30 years or older. Additionally, rs5015480 and rs9939609 were significantly associated with GDM in the same age group. These SNPs may therefore be more closely linked to GDM in older mothers.
Collapse
Affiliation(s)
- Qiaoli Zeng
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
- Key Laboratory of Research in Maternal and Child Medicine and Birth Defects, Guangdong Medical University, Foshan, Guangdong, China
- Matenal and Child Research Institute, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Jia Liu
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
- Key Laboratory of Research in Maternal and Child Medicine and Birth Defects, Guangdong Medical University, Foshan, Guangdong, China
- Matenal and Child Research Institute, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Xin Liu
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
- Key Laboratory of Research in Maternal and Child Medicine and Birth Defects, Guangdong Medical University, Foshan, Guangdong, China
- Matenal and Child Research Institute, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Na Liu
- Department of Pediatrics, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Weibiao Wu
- Medical Genetics Laboratory, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Ray Gyan Watson
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
- Key Laboratory of Research in Maternal and Child Medicine and Birth Defects, Guangdong Medical University, Foshan, Guangdong, China
- Matenal and Child Research Institute, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Dehua Zou
- School of Pharmacy, Macau University of Science and Technology, Macao, Macao SAR, China
- Guangdong Engineering Research Center of Chinese Medicine and Disease Susceptibility, Jinan University, Guangzhou, Guangdong, China
| | - Yue Wei
- Department of Ultrasound, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| | - Runmin Guo
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
- Key Laboratory of Research in Maternal and Child Medicine and Birth Defects, Guangdong Medical University, Foshan, Guangdong, China
- Matenal and Child Research Institute, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| |
Collapse
|
21
|
Yang Z, Kirschke CP, Cai Y, Huang L. A double knockout for zinc transporter 8 and somatostatin in mice reveals their distinct roles in regulation of insulin secretion and obesity. GENES & NUTRITION 2024; 19:24. [PMID: 39567934 PMCID: PMC11580226 DOI: 10.1186/s12263-024-00759-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/30/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Both zinc transporter 8 (ZnT8) and somatostatin (Sst) play crucial roles in the regulation of insulin and glucagon secretion. However, the interaction between them in controlling glucose metabolism was not well understood. The aim of this study was to explore the interactive effects of a double knockout of Znt8 and Sst on insulin and glucose metabolism in mice. METHODS Co-expression of ZnT8 with hormones secreted from gastrointestinal endocrine cells of mice was determined using immunofluorescence. Male Znt8 knockout (Znt8KO), Sst knockout (SstKO), double knockout for Sst and Znt8 (DKO), and the wild-type (WT) mice were fed a regular chow diet (CD) or a high-fat diet (HFD) at 3 weeks old for 15 weeks. Weights and fasting or fed glucose levels were determined. Glucose and insulin tolerance tests were performed; metabolic-relevant hormone levels including insulin, glucagon, glucagon-like peptide 1, Pyy, and leptin were determined. RESULTS ZnT8 is co-expressed with Sst in a subpopulation of endocrine D cells in the gastrointestinal tract. The absence of ZnT8 expression resulted in an increased density of the dense cores in the secretory granules of the D cell. DKO mice had reduced weight compared to WT when maintained on the CD. Compared to Znt8KO and SstKO, DKO mice did not show significant differences in fed or fasting blood glucose level regardless of dietary conditions. However, the CD-fed DKO mice had impaired insulin secretion without alterations in islet morphology or numbers. Moreover, DKO mice displayed diet-induced insulin resistance and disrupted secretion of metabolic-related hormones. CONCLUSIONS Somatostatin as well as a normal insulin sensitivity are required for normalizing glucose metabolism in Znt8KO mice. ZnT8 may play a role in regulating fat mass and leptin secretion. These findings shed light on the multifaceted nature of Znt8 and Sst interactions, opening new avenues to understand their roles in controlling glucose metabolism and fat mass.
Collapse
Affiliation(s)
- Zhongyue Yang
- Graduate Group of Nutritional Biology, Department of Nutrition, University of California at Davis, One Shields Ave, Davis, CA, 95616, USA
| | - Catherine P Kirschke
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA
| | - Yimeng Cai
- Graduate Group of Nutritional Biology, Department of Nutrition, University of California at Davis, One Shields Ave, Davis, CA, 95616, USA
| | - Liping Huang
- Graduate Group of Nutritional Biology, Department of Nutrition, University of California at Davis, One Shields Ave, Davis, CA, 95616, USA.
- USDA/ARS/Western Human Nutrition Research Center, 430 West Health Sciences Drive, Davis, CA, 95616, USA.
- Integrative Genetics and Genomics, University of California at Davis, One Shields Ave, Davis, CA, 95616, USA.
| |
Collapse
|
22
|
Imamura M, Maeda S. Genetic studies of type 2 diabetes, and microvascular complications of diabetes. Diabetol Int 2024; 15:699-706. [PMID: 39469559 PMCID: PMC11512959 DOI: 10.1007/s13340-024-00727-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/24/2024] [Indexed: 10/30/2024]
Abstract
Genome-wide association studies (GWAS) have significantly advanced the identification of genetic susceptibility variants associated with complex diseases. As of 2023, approximately 800 variants predisposing individuals to the risk of type 2 diabetes (T2D) were identified through GWAS, and the majority of studies were predominantly conducted in European populations. Despite the shared nature of the majority of genetic susceptibility loci across diverse ethnic populations, GWAS in non-European populations, including Japanese and East Asian populations, have revealed population-specific T2D loci. Currently, polygenic risk scores (PRSs), encompassing millions of associated variants, can identify individuals with a higher T2D risk than the general population. However, GWAS focusing on microvascular complications of diabetes have identified a limited number of disease-susceptibility loci. Ongoing efforts are crucial to enhance the applicability of PRS for all ethnic groups and unravel the genetic architecture of microvascular complications of diabetes.
Collapse
Affiliation(s)
- Minako Imamura
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara-Cho, Okinawa 903-0215 Japan
- Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Nishihara-Cho, Okinawa 930-0215 Japan
| | - Shiro Maeda
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara-Cho, Okinawa 903-0215 Japan
- Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Nishihara-Cho, Okinawa 930-0215 Japan
| |
Collapse
|
23
|
Shaked O, Loza BL, Olthoff KM, Reddy KR, Keating BJ, Testa G, Asrani SK, Shaked A. Donor and recipient genetics: Implications for the development of posttransplant diabetes mellitus. Am J Transplant 2024; 24:1794-1802. [PMID: 38782187 DOI: 10.1016/j.ajt.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/16/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
Posttransplant diabetes mellitus (PTDM) is a prevalent complication of liver transplantation and is associated with cardiometabolic complications. We studied the consequences of genetic effects of liver donors and recipients on PTDM outcomes, focusing on the diverse genetic pathways related to insulin that play a role in the development of PTDM. One thousand one hundred fifteen liver transplant recipients without a pretransplant diagnosis of type 2 diabetes mellitus (T2D) and their paired donors recruited from 2 transplant centers had polygenic risk scores (PRS) for T2D, insulin secretion, and insulin sensitivity calculated. Among recipients in the highest T2D-PRS quintile, donor T2D-PRS did not contribute significantly to PTDM. However, in recipients with the lowest T2D genetic risk, donor livers with the highest T2D-PRS contributed to the development of PTDM (OR [95% CI] = 3.79 [1.10-13.1], P = .035). Recipient risk was linked to factors associated with insulin secretion (OR [95% CI] = 0.85 [0.74-0.98], P = .02), while donor livers contributed to PTDM via gene pathways involved in insulin sensitivity (OR [95% CI] = 0.86 [0.75-0.99], P = .03). Recipient and donor PRS independently and collectively serve as predictors of PTDM onset. The genetically influenced biological pathways in recipients primarily pertain to insulin secretion, whereas the genetic makeup of donors exerts an influence on insulin sensitivity.
Collapse
Affiliation(s)
- Oren Shaked
- Department of Surgery, Penn Transplant Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bao-Li Loza
- Department of Surgery, Penn Transplant Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kim M Olthoff
- Department of Surgery, Penn Transplant Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kuchikula Rajender Reddy
- Department of Surgery, Penn Transplant Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brendan J Keating
- Department of Surgery, Penn Transplant Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Giuliano Testa
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX, USA
| | - Sumeet K Asrani
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX, USA
| | - Abraham Shaked
- Department of Surgery, Penn Transplant Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
24
|
Wu T, Hu Y, Tang LV. Gene therapy for polygenic or complex diseases. Biomark Res 2024; 12:99. [PMID: 39232780 PMCID: PMC11375922 DOI: 10.1186/s40364-024-00618-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
Gene therapy utilizes nucleic acid drugs to treat diseases, encompassing gene supplementation, gene replacement, gene silencing, and gene editing. It represents a distinct therapeutic approach from traditional medications and introduces novel strategies for genetic disorders. Over the past two decades, significant advancements have been made in the field of gene therapy, leading to the approval of various gene therapy drugs. Gene therapy was initially employed for treating genetic diseases and cancers, particularly monogenic conditions classified as orphan diseases due to their low prevalence rates; however, polygenic or complex diseases exhibit higher incidence rates within populations. Extensive research on the etiology of polygenic diseases has unveiled new therapeutic targets that offer fresh opportunities for their treatment. Building upon the progress achieved in gene therapy for monogenic diseases and cancers, extending its application to polygenic or complex diseases would enable targeting a broader range of patient populations. This review aims to discuss the strategies of gene therapy, methods of gene editing (mainly CRISPR-CAS9), and carriers utilized in gene therapy, and highlight the applications of gene therapy in polygenic or complex diseases focused on applications that have either entered clinical stages or are currently undergoing clinical trials.
Collapse
Affiliation(s)
- Tingting Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| | - Liang V Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| |
Collapse
|
25
|
Nadia SN, Hasib M, Hasan I, Saba AA, Sayem M, Ebihara A, Hasan AM, Nabi AN. Genetic analyses of truncated variant rs200185429 in ZNT8 encoding SLC30A8 gene with respect to prediabetes and type 2 diabetes in Bangladeshi population. ENDOCRINE AND METABOLIC SCIENCE 2024; 16:100189. [DOI: 10.1016/j.endmts.2024.100189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
|
26
|
Díaz-García JD, Leyva-Leyva M, Sánchez-Aguillón F, de León-Bautista MP, Fuentes-Venegas A, Torres-Viloria A, Tenorio-Aguirre EK, Morales-Lázaro SL, Olivo-Díaz A, González-Ramírez R. Association Study of CACNA1D, KCNJ11, KCNQ1, and CACNA1E Single-Nucleotide Polymorphisms with Type 2 Diabetes Mellitus. Int J Mol Sci 2024; 25:9196. [PMID: 39273144 PMCID: PMC11395491 DOI: 10.3390/ijms25179196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a complex chronic disease characterized by decreased insulin secretion and the development of insulin resistance. Previous genome-wide association studies demonstrated that single-nucleotide polymorphisms (SNPs) present in genes coding for ion channels involved in insulin secretion increase the risk of developing this disease. We determined the association of 16 SNPs found in CACNA1D, KCNQ1, KCNJ11, and CACNA1E genes and the increased probability of developing T2DM. In this work, we performed a case-control study in 301 Mexican adults, including 201 cases with diabetes and 100 controls without diabetes. Our findings indicate a moderate association between T2DM and the C allele, and the C/C genotype of rs312480 within CACNA1D. The CAG haplotype surprisingly showed a protective effect, whereas the CAC and CGG haplotypes have a strong association with T2DM. The C allele and C/C genotype of rs5219 were significantly associated with diabetes. Also, an association was observed between diabetes and the A allele and the A/A genotype of rs3753737 and rs175338 in CACNA1E. The TGG and CGA haplotypes were also found to be significantly associated. The findings of this study indicate that the SNPs examined could serve as a potential diagnostic tool and contribute to the susceptibility of the Mexican population to this disease.
Collapse
Affiliation(s)
- Juan Daniel Díaz-García
- División de Medicina Interna, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (J.D.D.-G.); (A.F.-V.); (A.T.-V.); (E.K.T.-A.)
| | - Margarita Leyva-Leyva
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (M.L.-L.); (F.S.-A.); (A.O.-D.)
| | - Fabiola Sánchez-Aguillón
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (M.L.-L.); (F.S.-A.); (A.O.-D.)
| | - Mercedes Piedad de León-Bautista
- Escuela de Medicina, Universidad Vasco de Quiroga, Morelia 58090, Mexico;
- Laboratorio de Enfermedades Infecciosas y Genómica (INEX LAB), Morelia 58280, Mexico
| | - Abel Fuentes-Venegas
- División de Medicina Interna, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (J.D.D.-G.); (A.F.-V.); (A.T.-V.); (E.K.T.-A.)
| | - Alfredo Torres-Viloria
- División de Medicina Interna, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (J.D.D.-G.); (A.F.-V.); (A.T.-V.); (E.K.T.-A.)
| | - Erika Karina Tenorio-Aguirre
- División de Medicina Interna, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (J.D.D.-G.); (A.F.-V.); (A.T.-V.); (E.K.T.-A.)
| | - Sara Luz Morales-Lázaro
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
- Centro de Investigación Sobre el Envejecimiento, CINVESTAV, Mexico City 14330, Mexico
| | - Angélica Olivo-Díaz
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (M.L.-L.); (F.S.-A.); (A.O.-D.)
| | - Ricardo González-Ramírez
- Departamento de Biología Molecular e Histocompatibilidad, Hospital General “Dr. Manuel Gea González”, Mexico City 14080, Mexico; (M.L.-L.); (F.S.-A.); (A.O.-D.)
- Centro de Investigación Sobre el Envejecimiento, CINVESTAV, Mexico City 14330, Mexico
| |
Collapse
|
27
|
Rehn F, Kraemer-Schulien V, Bujnicki T, Bannach O, Tschoepe D, Stratmann B, Willbold D. IAPP - oligomerisation levels in plasma of people with type 2 diabetes. Sci Rep 2024; 14:19556. [PMID: 39174611 PMCID: PMC11341561 DOI: 10.1038/s41598-024-70255-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024] Open
Abstract
Islet amyloid polypeptide (IAPP) is co-secreted with insulin from pancreatic ß-cells. Its oligomerisation is regarded as disease driving force in type 2 diabetes (T2D) pathology. Up to now, IAPP oligomers have been detected in affected tissues. IAPP oligomer concentrations in blood have not been analysed so far. Using the IAPP single-oligomer-sensitive and monomer-insensitive surface-based fluorescence intensity distribution analysis (sFIDA) technology, levels of IAPP oligomers in blood plasma from healthy controls and people with T2D in different disease stages where determined. Subsequently, the level of IAPP oligomerisation was introduced as the ratio between the IAPP oligomers determined with sFIDA and the total IAPP concentration determined with ELISA. Highest oligomerisation levels were detected in plasma of people with T2D without late complication and without insulin therapy. Their levels stand out significantly from the control group. Healthy controls presented with the lowest oligomerisation levels in plasma. In people with T2D without complications, IAPP oligomerisation levels correlated with disease duration. The results clearly demonstrate that IAPP oligomerisation in insulin-naïve patients correlates with duration of T2D. Although a correlation per se does not identify, which is cause and what is consequence, this result supports the hypothesis that IAPP aggregation is the driving factor of T2D development and progression. The alternative and conventional hypothesis explains development of T2D with increasing insulin resistance causing exhaustion of pancreatic ß-cells due to over-secretion of insulin, and thus IAPP, too, resulting in subsequent IAPP aggregation and fibril deposition in the pancreas. Further experiments and comparative analyses with primary tissues are warranted.
Collapse
Affiliation(s)
- Fabian Rehn
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
- Institute of Biological Information Processing (Structural Biochemistry: IBI-7), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428, Jülich, Germany
- attyloid GmbH, Merowingerplatz 1A, 40225, Düsseldorf, Germany
| | - Victoria Kraemer-Schulien
- Institute of Biological Information Processing (Structural Biochemistry: IBI-7), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428, Jülich, Germany
| | - Tuyen Bujnicki
- Institute of Biological Information Processing (Structural Biochemistry: IBI-7), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428, Jülich, Germany
| | - Oliver Bannach
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
- Institute of Biological Information Processing (Structural Biochemistry: IBI-7), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428, Jülich, Germany
- attyloid GmbH, Merowingerplatz 1A, 40225, Düsseldorf, Germany
| | - Diethelm Tschoepe
- Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Medizinische Fakultät OWL (Universität Bielefeld), Georgstr. 11, 32545, Bad Oeynhausen, Germany
- Stiftung DHG (Diabetes I Herz I Gefäße) in der Deutschen Diabetes Stiftung, c/o Deutsches Diabetes-Zentrum (DDZ), Auf´m Hennekamp 65, 40225, Düsseldorf, Germany
| | - Bernd Stratmann
- Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Medizinische Fakultät OWL (Universität Bielefeld), Georgstr. 11, 32545, Bad Oeynhausen, Germany.
| | - Dieter Willbold
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany.
- Institute of Biological Information Processing (Structural Biochemistry: IBI-7), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428, Jülich, Germany.
- attyloid GmbH, Merowingerplatz 1A, 40225, Düsseldorf, Germany.
| |
Collapse
|
28
|
Meulebrouck S, Merrheim J, Queniat G, Bourouh C, Derhourhi M, Boissel M, Yi X, Badreddine A, Boutry R, Leloire A, Toussaint B, Amanzougarene S, Vaillant E, Durand E, Loiselle H, Huyvaert M, Dechaume A, Scherrer V, Marchetti P, Balkau B, Charpentier G, Franc S, Marre M, Roussel R, Scharfmann R, Cnop M, Canouil M, Baron M, Froguel P, Bonnefond A. Functional genetics reveals the contribution of delta opioid receptor to type 2 diabetes and beta-cell function. Nat Commun 2024; 15:6627. [PMID: 39103322 PMCID: PMC11300616 DOI: 10.1038/s41467-024-51004-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024] Open
Abstract
Functional genetics has identified drug targets for metabolic disorders. Opioid use impacts metabolic homeostasis, although mechanisms remain elusive. Here, we explore the OPRD1 gene (encoding delta opioid receptor, DOP) to understand its impact on type 2 diabetes. Large-scale sequencing of OPRD1 and in vitro analysis reveal that loss-of-function variants are associated with higher adiposity and lower hyperglycemia risk, whereas gain-of-function variants are associated with lower adiposity and higher type 2 diabetes risk. These findings align with studies of opium addicts. OPRD1 is expressed in human islets and beta cells, with decreased expression under type 2 diabetes conditions. DOP inhibition by an antagonist enhances insulin secretion from human beta cells and islets. RNA-sequencing identifies pathways regulated by DOP antagonism, including nerve growth factor, circadian clock, and nuclear receptor pathways. Our study highlights DOP as a key player between opioids and metabolic homeostasis, suggesting its potential as a therapeutic target for type 2 diabetes.
Collapse
Grants
- This study was funded by the French National Research Agency (ANR-10-LABX-46 [European Genomics Institute for Diabetes] to PF and AB), the French National Research Agency (ANR-10-EQPX-07-01 [LIGAN-PM] to PF and AB), the European Research Council (ERC Reg-Seq – 715575 and ERC OpiO – 101043671, to AB), the EFSD New Targets for Diabetes or Obesity-related Metabolic Diseases Programme supported by an educational research grant from MSD (to AB) and the National Center for Precision Diabetic Medicine – PreciDIAB, which is jointly supported by the French National Agency for Research (ANR-18-IBHU-0001), by the European Union (FEDER), by the Hauts-de-France Regional Council and by the European Metropolis of Lille (MEL). The study was also supported by "France Génomique" consortium (ANR-10-INBS-009). XY was supported by the Fondation ULB and the China Scholarship Council. MCnop acknowledges support by the Walloon Region SPW-EER (Win2Wal project BetaSource), the Fonds National de la Recherche Scientifique (FRS-FNRS) and the Francophone Foundation for Diabetes Research (FFRD, that is sponsored by the French Diabetes Federation, Abbott, Eli Lilly, Merck Sharp & Dohme and Novo Nordisk).
Collapse
Affiliation(s)
- Sarah Meulebrouck
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Judith Merrheim
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Gurvan Queniat
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Cyril Bourouh
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Mehdi Derhourhi
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Mathilde Boissel
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Xiaoyan Yi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Alaa Badreddine
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Raphaël Boutry
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Audrey Leloire
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Bénédicte Toussaint
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Souhila Amanzougarene
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Emmanuel Vaillant
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Emmanuelle Durand
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Hélène Loiselle
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Marlène Huyvaert
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Aurélie Dechaume
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Victoria Scherrer
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Piero Marchetti
- Islet Cell Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Beverley Balkau
- Paris-Saclay University, Paris-Sud University, UVSQ, Center for Research in Epidemiology and Population Health, Inserm U1018 Clinical Epidemiology, Villejuif, France
| | - Guillaume Charpentier
- CERITD (Centre d'Étude et de Recherche pour l'Intensification du Traitement du Diabète), Evry, France
| | - Sylvia Franc
- CERITD (Centre d'Étude et de Recherche pour l'Intensification du Traitement du Diabète), Evry, France
- Department of Diabetes, Sud-Francilien Hospital, Paris-Sud University, Corbeil-Essonnes, France
| | - Michel Marre
- Institut Necker-Enfants Malades, Inserm, Université de Paris, Paris, France
- Clinique Ambroise Paré, Neuilly-sur-Seine, France
| | - Ronan Roussel
- Institut Necker-Enfants Malades, Inserm, Université de Paris, Paris, France
- Department of Diabetology Endocrinology Nutrition, Hôpital Bichat, DHU FIRE, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Raphaël Scharfmann
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université de Paris, Paris, France
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
- Division of Endocrinology, ULB Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Mickaël Canouil
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Morgane Baron
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Philippe Froguel
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France.
- Department of Metabolism, Imperial College London, London, UK.
| | - Amélie Bonnefond
- Université de Lille, Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France.
- Department of Metabolism, Imperial College London, London, UK.
| |
Collapse
|
29
|
Nammo T, Funahashi N, Udagawa H, Kozawa J, Nakano K, Shimizu Y, Okamura T, Kawaguchi M, Uebanso T, Nishimura W, Hiramoto M, Shimomura I, Yasuda K. Single-housing-induced islet epigenomic changes are related to polymorphisms in diabetic KK mice. Life Sci Alliance 2024; 7:e202302099. [PMID: 38876803 PMCID: PMC11178941 DOI: 10.26508/lsa.202302099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/16/2024] Open
Abstract
A lack of social relationships is increasingly recognized as a type 2 diabetes (T2D) risk. To investigate the underlying mechanism, we used male KK mice, an inbred strain with spontaneous diabetes. Given the association between living alone and T2D risk in humans, we divided the non-diabetic mice into singly housed (KK-SH) and group-housed control mice. Around the onset of diabetes in KK-SH mice, we compared H3K27ac ChIP-Seq with RNA-Seq using pancreatic islets derived from each experimental group, revealing a positive correlation between single-housing-induced changes in H3K27ac and gene expression levels. In particular, single-housing-induced H3K27ac decreases revealed a significant association with islet cell functions and GWAS loci for T2D and related diseases, with significant enrichment of binding motifs for transcription factors representative of human diabetes. Although these H3K27ac regions were preferentially localized to a polymorphic genomic background, SNVs and indels did not cause sequence disruption of enriched transcription factor motifs in most of these elements. These results suggest alternative roles of genetic variants in environment-dependent epigenomic changes and provide insights into the complex mode of disease inheritance.
Collapse
Affiliation(s)
- Takao Nammo
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Diabetes Care Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Nobuaki Funahashi
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Haruhide Udagawa
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Registered Dietitians, Faculty of Health and Nutrition, Bunkyo University, Chigasaki, Japan
| | - Junji Kozawa
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Diabetes Care Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kenta Nakano
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan
| | - Yukiko Shimizu
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, Japan
| | - Miho Kawaguchi
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Takashi Uebanso
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Preventive Environment and Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Wataru Nishimura
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Molecular Biology, International University of Health and Welfare School of Medicine, Chiba, Japan
- Division of Anatomy, Bio-Imaging and Neuro-cell Science, Jichi Medical University, Tochigi, Japan
| | - Masaki Hiramoto
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Biochemistry, Tokyo Medical University, Tokyo, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuki Yasuda
- Department of Metabolic Disorder, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Diabetes, Endocrinology and Metabolism, Kyorin University School of Medicine, Tokyo, Japan
| |
Collapse
|
30
|
Camaya I, Hill M, Sais D, Tran N, O'Brien B, Donnelly S. The Parasite-Derived Peptide, FhHDM-1, Selectively Modulates miRNA Expression in β-Cells to Prevent Apoptotic Pathways Induced by Proinflammatory Cytokines. J Diabetes Res 2024; 2024:8555211. [PMID: 39022651 PMCID: PMC11254460 DOI: 10.1155/2024/8555211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/04/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
We have previously identified a parasite-derived peptide, FhHDM-1, that prevented the progression of diabetes in nonobese diabetic (NOD) mice. Disease prevention was mediated by the activation of the PI3K/Akt pathway to promote β-cell survival and metabolism without inducing proliferation. To determine the molecular mechanisms driving the antidiabetogenic effects of FhHDM-1, miRNA:mRNA interactions and in silico predictions of the gene networks were characterised in β-cells, which were exposed to the proinflammatory cytokines that mediate β-cell destruction in Type 1 diabetes (T1D), in the presence and absence of FhHDM-1. The predicted gene targets of miRNAs differentially regulated by FhHDM-1 mapped to the biological pathways that regulate β-cell biology. Six miRNAs were identified as important nodes in the regulation of PI3K/Akt signaling. Additionally, IGF-2 was identified as a miRNA gene target that mediated the beneficial effects of FhHDM-1 on β-cells. The findings provide a putative mechanism by which FhHDM-1 positively impacts β-cells to permanently prevent diabetes. As β-cell death/dysfunction underlies diabetes development, FhHDM-1 opens new therapeutic avenues.
Collapse
Affiliation(s)
- Inah Camaya
- The School of Life SciencesUniversity of Technology Sydney, Ultimo, New South Wales, Australia
| | - Meredith Hill
- School of Biomedical EngineeringFaculty of Engineering and Information TechnologyUniversity of Technology Sydney, Ultimo, New South Wales, Australia
| | - Dayna Sais
- School of Biomedical EngineeringFaculty of Engineering and Information TechnologyUniversity of Technology Sydney, Ultimo, New South Wales, Australia
| | - Nham Tran
- School of Biomedical EngineeringFaculty of Engineering and Information TechnologyUniversity of Technology Sydney, Ultimo, New South Wales, Australia
| | - Bronwyn O'Brien
- The School of Life SciencesUniversity of Technology Sydney, Ultimo, New South Wales, Australia
| | - Sheila Donnelly
- The School of Life SciencesUniversity of Technology Sydney, Ultimo, New South Wales, Australia
| |
Collapse
|
31
|
Digles D, Ingles-Prieto A, Dvorak V, Mocking TAM, Goldmann U, Garofoli A, Homan EJ, Di Silvio A, Azzollini L, Sassone F, Fogazza M, Bärenz F, Pommereau A, Zuschlag Y, Ooms JF, Tranberg-Jensen J, Hansen JS, Stanka J, Sijben HJ, Batoulis H, Bender E, Martini R, IJzerman AP, Sauer DB, Heitman LH, Manolova V, Reinhardt J, Ehrmann A, Leippe P, Ecker GF, Huber KVM, Licher T, Scarabottolo L, Wiedmer T, Superti-Furga G. Advancing drug discovery through assay development: a survey of tool compounds within the human solute carrier superfamily. Front Pharmacol 2024; 15:1401599. [PMID: 39050757 PMCID: PMC11267547 DOI: 10.3389/fphar.2024.1401599] [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: 03/15/2024] [Accepted: 05/31/2024] [Indexed: 07/27/2024] Open
Abstract
With over 450 genes, solute carriers (SLCs) constitute the largest transporter superfamily responsible for the uptake and efflux of nutrients, metabolites, and xenobiotics in human cells. SLCs are associated with a wide variety of human diseases, including cancer, diabetes, and metabolic and neurological disorders. They represent an important therapeutic target class that remains only partly exploited as therapeutics that target SLCs are scarce. Additionally, many small molecules reported in the literature to target SLCs are poorly characterized. Both features may be due to the difficulty of developing SLC transport assays that fulfill the quality criteria for high-throughput screening. Here, we report one of the main limitations hampering assay development within the RESOLUTE consortium: the lack of a resource providing high-quality information on SLC tool compounds. To address this, we provide a systematic annotation of tool compounds targeting SLCs. We first provide an overview on RESOLUTE assays. Next, we present a list of SLC-targeting compounds collected from the literature and public databases; we found that most data sources lacked specificity data. Finally, we report on experimental tests of 19 selected compounds against a panel of 13 SLCs from seven different families. Except for a few inhibitors, which were active on unrelated SLCs, the tested inhibitors demonstrated high selectivity for their reported targets. To make this knowledge easily accessible to the scientific community, we created an interactive dashboard displaying the collected data in the RESOLUTE web portal (https://re-solute.eu). We anticipate that our open-access resources on assays and compounds will support the development of future drug discovery campaigns for SLCs.
Collapse
Affiliation(s)
- Daniela Digles
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Alvaro Ingles-Prieto
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Vojtech Dvorak
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Tamara A. M. Mocking
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, Netherlands
| | - Ulrich Goldmann
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Andrea Garofoli
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Evert J. Homan
- Science for Life Laboratory, Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | - Felix Bärenz
- Sanofi, Integrated Drug Discovery, Industriepark Hoechst, Frankfurt am Main, Hessen, Germany
| | - Antje Pommereau
- Sanofi, Integrated Drug Discovery, Industriepark Hoechst, Frankfurt am Main, Hessen, Germany
| | - Yasmin Zuschlag
- Sanofi, Integrated Drug Discovery, Industriepark Hoechst, Frankfurt am Main, Hessen, Germany
| | - Jasper F. Ooms
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, Netherlands
| | - Jeppe Tranberg-Jensen
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jesper S. Hansen
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Josefina Stanka
- Lead Identification and Characterization, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Hubert J. Sijben
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, Netherlands
| | - Helena Batoulis
- Lead Identification and Characterization, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Eckhard Bender
- Lead Identification and Characterization, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Riccardo Martini
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Adriaan P. IJzerman
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, Netherlands
| | - David B. Sauer
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Laura H. Heitman
- Division of Drug Discovery and Safety, LACDR, Leiden University, Leiden, Netherlands
| | | | | | - Alexander Ehrmann
- Lead Identification and Characterization, Bayer Pharmaceuticals, Wuppertal, Germany
| | - Philipp Leippe
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Gerhard F. Ecker
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Kilian V. M. Huber
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Thomas Licher
- Sanofi, Integrated Drug Discovery, Industriepark Hoechst, Frankfurt am Main, Hessen, Germany
| | | | - Tabea Wiedmer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
32
|
Ran J, Guo G, Zhang S, Zhang Y, Zhang L, Li D, Wu S, Cong Y, Wang X, Xie S, Zhao H, Liu H, Ou G, Zhu X, Zhou J, Liu M. KIF11 UFMylation Maintains Photoreceptor Cilium Integrity and Retinal Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400569. [PMID: 38666385 PMCID: PMC11220646 DOI: 10.1002/advs.202400569] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/01/2024] [Indexed: 07/04/2024]
Abstract
The photoreceptor cilium is vital for maintaining the structure and function of the retina. However, the molecular mechanisms underlying the photoreceptor cilium integrity and retinal homeostasis are largely unknown. Herein, it is shown that kinesin family member 11 (KIF11) localizes at the transition zone (connecting cilium) of the photoreceptor and plays a crucial role in orchestrating the cilium integrity. KIF11 depletion causes malformations of both the photoreceptor ciliary axoneme and membranous discs, resulting in photoreceptor degeneration and the accumulation of drusen-like deposits throughout the retina. Mechanistic studies show that the stability of KIF11 is regulated by an interplay between its UFMylation and ubiquitination; UFMylation of KIF11 at lysine 953 inhibits its ubiquitination by synoviolin 1 and thereby prevents its proteasomal degradation. The lysine 953-to-arginine mutant of KIF11 is more stable than wild-type KIF11 and also more effective in reversing the ciliary and retinal defects induced by KIF11 depletion. These findings identify a critical role for KIF11 UFMylation in the maintenance of photoreceptor cilium integrity and retinal homeostasis.
Collapse
Affiliation(s)
- Jie Ran
- Center for Cell Structure and FunctionShandong Provincial Key Laboratory of Animal Resistance BiologyHaihe Laboratory of Cell EcosystemCollege of Life SciencesShandong Normal UniversityJinan250014China
| | - Guizhi Guo
- Center for Cell Structure and FunctionShandong Provincial Key Laboratory of Animal Resistance BiologyHaihe Laboratory of Cell EcosystemCollege of Life SciencesShandong Normal UniversityJinan250014China
| | - Sai Zhang
- Center for Cell Structure and FunctionShandong Provincial Key Laboratory of Animal Resistance BiologyHaihe Laboratory of Cell EcosystemCollege of Life SciencesShandong Normal UniversityJinan250014China
| | - Yufei Zhang
- Center for Cell Structure and FunctionShandong Provincial Key Laboratory of Animal Resistance BiologyHaihe Laboratory of Cell EcosystemCollege of Life SciencesShandong Normal UniversityJinan250014China
| | - Liang Zhang
- Center for Cell Structure and FunctionShandong Provincial Key Laboratory of Animal Resistance BiologyHaihe Laboratory of Cell EcosystemCollege of Life SciencesShandong Normal UniversityJinan250014China
| | - Dengwen Li
- Department of Genetics and Cell BiologyState Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjin300071China
| | - Shian Wu
- Department of Genetics and Cell BiologyState Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjin300071China
| | - Yusheng Cong
- Key Laboratory of Aging and Cancer Biology of Zhejiang ProvinceInstitute of Aging ResearchSchool of MedicineHangzhou Normal UniversityHangzhou310036China
| | - Xiaohong Wang
- Department of PharmacologyTianjin Key Laboratory of Inflammation BiologySchool of Basic Medical SciencesTianjin Medical UniversityTianjin300070China
| | - Songbo Xie
- Center for Cell Structure and FunctionShandong Provincial Key Laboratory of Animal Resistance BiologyHaihe Laboratory of Cell EcosystemCollege of Life SciencesShandong Normal UniversityJinan250014China
| | - Huijie Zhao
- Center for Cell Structure and FunctionShandong Provincial Key Laboratory of Animal Resistance BiologyHaihe Laboratory of Cell EcosystemCollege of Life SciencesShandong Normal UniversityJinan250014China
| | - Hongbin Liu
- Center for Reproductive MedicineCheeloo College of MedicineKey Laboratory of Reproductive Endocrinology of Ministry of EducationShandong UniversityJinan250014China
| | - Guangshuo Ou
- Tsinghua‐Peking Center for Life SciencesMinistry of Education Key Laboratory for Protein ScienceSchool of Life SciencesTsinghua UniversityBeijing100084China
| | - Xueliang Zhu
- State Key Laboratory of Cell BiologyCAS Center for Excellence in Molecular Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghai200031China
| | - Jun Zhou
- Center for Cell Structure and FunctionShandong Provincial Key Laboratory of Animal Resistance BiologyHaihe Laboratory of Cell EcosystemCollege of Life SciencesShandong Normal UniversityJinan250014China
- Department of Genetics and Cell BiologyState Key Laboratory of Medicinal Chemical BiologyCollege of Life SciencesNankai UniversityTianjin300071China
| | - Min Liu
- Laboratory of Tissue HomeostasisHaihe Laboratory of Cell EcosystemTianjin300462China
| |
Collapse
|
33
|
Azzollini L, Prete DD, Wolf G, Klimek C, Saggioro M, Ricci F, Christodoulaki E, Wiedmer T, Ingles-Prieto A, Superti-Furga G, Scarabottolo L. Development of a live cell assay for the zinc transporter ZnT8. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100166. [PMID: 38848895 DOI: 10.1016/j.slasd.2024.100166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/27/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Zinc is an essential trace element that is involved in many biological processes and in cellular homeostasis. In pancreatic β-cells, zinc is crucial for the synthesis, processing, and secretion of insulin, which plays a key role in glucose homeostasis and which deficiency is the cause of diabetes. The accumulation of zinc in pancreatic cells is regulated by the solute carrier transporter SLC30A8 (or Zinc Transporter 8, ZnT8), which transports zinc from cytoplasm in intracellular vesicles. Allelic variants of SLC30A8 gene have been linked to diabetes. Given the physiological intracellular localization of SLC30A8 in pancreatic β-cells and the ubiquitous endogenous expression of other Zinc transporters in different cell lines that could be used as cellular model for SLC30A8 recombinant over-expression, it is challenging to develop a functional assay to measure SLC30A8 activity. To achieve this goal, we have firstly generated a HEK293 cell line stably overexpressing SLC30A8, where the over-expression favors the partial localization of SLC30A8 on the plasma membrane. Then, we used the combination of this cell model, commercial FluoZin-3 cell permeant zinc dye and live cell imaging approach to follow zinc flux across SLC30A8 over-expressed on plasma membrane, thus developing a novel functional imaging- based assay specific for SLC30A8. Our novel approach can be further explored and optimized, paving the way for future small molecule medium-throughput screening.
Collapse
Affiliation(s)
- Lucia Azzollini
- Axxam SpA, Openzone, Via Meucci 3 20091 Bresso, Milan, Italy.
| | | | - Gernot Wolf
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Christoph Klimek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Mattia Saggioro
- Axxam SpA, Openzone, Via Meucci 3 20091 Bresso, Milan, Italy
| | - Fernanda Ricci
- Axxam SpA, Openzone, Via Meucci 3 20091 Bresso, Milan, Italy
| | - Eirini Christodoulaki
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Tabea Wiedmer
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Alvaro Ingles-Prieto
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
34
|
Kim DS, Song L, Gou W, Kim J, Liu B, Wei H, Muise-Helmericks RC, Li Z, Wang H. GRP94 is an IGF-1R chaperone and regulates beta cell death in diabetes. Cell Death Dis 2024; 15:374. [PMID: 38811543 PMCID: PMC11137047 DOI: 10.1038/s41419-024-06754-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
High workload-induced cellular stress can cause pancreatic islet β cell death and dysfunction, or β cell failure, a hallmark of type 2 diabetes mellitus. Thus, activation of molecular chaperones and other stress-response genes prevents β cell failure. To this end, we have shown that deletion of the glucose-regulated protein 94 (GRP94) in Pdx1+ pancreatic progenitor cells led to pancreas hypoplasia and reduced β cell mass during pancreas development in mice. Here, we show that GRP94 was involved in β cell adaption and compensation (or failure) in islets from leptin receptor-deficient (db/db) mice in an age-dependent manner. GRP94-deficient cells were more susceptible to cell death induced by various diabetogenic stress conditions. We also identified a new client of GRP94, insulin-like growth factor-1 receptor (IGF-1R), a critical factor for β cell survival and function that may mediate the effect of GRP94 in the pathogenesis of diabetes. This study has identified essential functions of GRP94 in β cell failure related to diabetes.
Collapse
Affiliation(s)
- Do-Sung Kim
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Lili Song
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Wenyu Gou
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Jisun Kim
- Microbiology and Immunology, Medical University of South Carolina, Charleson, SC, 29425, USA
| | - Bei Liu
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-James, Columbus, OH, 43210, USA
| | - Hua Wei
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Robin C Muise-Helmericks
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-James, Columbus, OH, 43210, USA
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.
| |
Collapse
|
35
|
Zhao Y, Ansarullah, Kumar P, Mahoney JM, He H, Baker C, George J, Li S. Causal network perturbation analysis identifies known and novel type-2 diabetes driver genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595431. [PMID: 38826370 PMCID: PMC11142180 DOI: 10.1101/2024.05.22.595431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The molecular pathogenesis of diabetes is multifactorial, involving genetic predisposition and environmental factors that are not yet fully understood. However, pancreatic β-cell failure remains among the primary reasons underlying the progression of type-2 diabetes (T2D) making targeting β-cell dysfunction an attractive pathway for diabetes treatment. To identify genetic contributors to β-cell dysfunction, we investigated single-cell gene expression changes in β-cells from healthy (C57BL/6J) and diabetic (NZO/HlLtJ) mice fed with normal or high-fat, high-sugar diet (HFHS). Our study presents an innovative integration of the causal network perturbation assessment (ssNPA) framework with meta-cell transcriptome analysis to explore the genetic underpinnings of type-2 diabetes (T2D). By generating a reference causal network and in silico perturbation, we identified novel genes implicated in T2D and validated our candidates using the Knockout Mouse Phenotyping (KOMP) Project database.
Collapse
Affiliation(s)
- Yue Zhao
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Ansarullah
- Center for Biometric Analysis, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Parveen Kumar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Hao He
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Candice Baker
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Joshy George
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Sheng Li
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
36
|
Wang SH, Huang YC, Cheng CW, Chang YW, Liao WL. Impact of the trans-ancestry polygenic risk score on type 2 diabetes risk, onset age and progression among population in Taiwan. Am J Physiol Endocrinol Metab 2024; 326:E547-E554. [PMID: 38363735 PMCID: PMC11376485 DOI: 10.1152/ajpendo.00252.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 02/18/2024]
Abstract
Type 2 diabetes (T2D) prevalence in adults at a younger age has increased but the disease status may go unnoticed. This study aimed to determine whether the onset age and subsequent diabetic complications can be attributed to the polygenic architecture of T2D in the Taiwan Han population. A total of 9,627 cases with T2D and 85,606 controls from the Taiwan Biobank were enrolled. Three diabetic polygenic risk scores (PRSs), PRS_EAS and PRS_EUR, and a trans-ancestry PRS (PRS_META), calculated using summary statistic from East Asian and European populations. The onset age was identified by linking to the National Taiwan Insurance Research Database, and the incidence of different diabetic complications during follow-up was recorded. PRS_META (7.4%) explained a higher variation for T2D status. And the higher percentile of PRS is also correlated with higher percentage of T2D family history and prediabetes status. More, the PRS was negatively associated with onset age (β = -0.91 yr), and this was more evident among males (β = -1.11 vs. -0.76 for males and females, respectively). The hazard ratio of diabetic retinopathy (DR) and diabetic foot were significantly associated with PRS_EAS and PRS_META, respectively. However, the PRS was not associated with other diabetic complications, including diabetic nephropathy, cardiovascular disease, and hypertension. Our findings indicated that diabetic PRS which combined susceptibility variants from cross-population could be used as a tool for early screening of T2D, especially for high-risk populations, such as individuals with high genetic risk, and may be associated with the risk of complications in subjects with T2D. NEW & NOTEWORTHY Our findings indicated that diabetic polygenic risk score (PRS) which combined susceptibility variants from Asian and European population affect the onset age of type 2 diabetes (T2D) and could be used as a tool for early screening of T2D, especially for individuals with high genetic risk, and may be associated with the risk of diabetic complications among people in Taiwan.
Collapse
Affiliation(s)
- Shi-Heng Wang
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Zhunan, Taiwan
- Department of Public Health, China Medical University, Taichung, Taiwan
| | - Yu-Chuen Huang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Chun-Wen Cheng
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ya-Wen Chang
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Center for Personalized Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Ling Liao
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Center for Personalized Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
37
|
Hu M, Kim I, Morán I, Peng W, Sun O, Bonnefond A, Khamis A, Bonàs-Guarch S, Froguel P, Rutter GA. Multiple genetic variants at the SLC30A8 locus affect local super-enhancer activity and influence pancreatic β-cell survival and function. FASEB J 2024; 38:e23610. [PMID: 38661000 PMCID: PMC11108099 DOI: 10.1096/fj.202301700rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/22/2024] [Accepted: 04/01/2024] [Indexed: 04/26/2024]
Abstract
Variants at the SLC30A8 locus are associated with type 2 diabetes (T2D) risk. The lead variant, rs13266634, encodes an amino acid change, Arg325Trp (R325W), at the C-terminus of the secretory granule-enriched zinc transporter, ZnT8. Although this protein-coding variant was previously thought to be the sole driver of T2D risk at this locus, recent studies have provided evidence for lowered expression of SLC30A8 mRNA in protective allele carriers. In the present study, we examined multiple variants that influence SLC30A8 allele-specific expression. Epigenomic mapping has previously identified an islet-selective enhancer cluster at the SLC30A8 locus, hosting multiple T2D risk and cASE associations, which is spatially associated with the SLC30A8 promoter and additional neighboring genes. Here, we show that deletion of variant-bearing enhancer regions using CRISPR-Cas9 in human-derived EndoC-βH3 cells lowers the expression of SLC30A8 and several neighboring genes and improves glucose-stimulated insulin secretion. While downregulation of SLC30A8 had no effect on beta cell survival, loss of UTP23, RAD21, or MED30 markedly reduced cell viability. Although eQTL or cASE analyses in human islets did not support the association between these additional genes and diabetes risk, the transcriptional regulator JQ1 lowered the expression of multiple genes at the SLC30A8 locus and enhanced stimulated insulin secretion.
Collapse
Affiliation(s)
- Ming Hu
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Innah Kim
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Ignasi Morán
- Life Sciences Department, Barcelona Supercomputing Center (BSC-CNS), 08034 Barcelona, Spain
| | - Weicong Peng
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Orien Sun
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Amélie Bonnefond
- Department of Metabolism, Digestion, and Reproduction, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
- Inserm U1283, CNRS UMR 8199, EGID, Institut Pasteur de Lille, F-59000, France
- University of Lille, Lille University Hospital, Lille, F-59000, France.France
| | - Amna Khamis
- Department of Metabolism, Digestion, and Reproduction, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
- Inserm U1283, CNRS UMR 8199, EGID, Institut Pasteur de Lille, F-59000, France
- University of Lille, Lille University Hospital, Lille, F-59000, France.France
| | - Sílvia Bonàs-Guarch
- Department of Metabolism, Digestion, and Reproduction, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
- Center for Genomic Regulation (CRG), C/ Dr. Aiguader, 88, PRBB Building, 08003 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Philippe Froguel
- Department of Metabolism, Digestion, and Reproduction, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
- Inserm U1283, CNRS UMR 8199, EGID, Institut Pasteur de Lille, F-59000, France
- University of Lille, Lille University Hospital, Lille, F-59000, France.France
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
- Centre de Recherche du CHUM, Faculté de Médicine, Université de Montréal, Montréal, QC, Canada
- Lee Kong Chian Imperial Medical School, Nanyang Technological University, Singapore
| |
Collapse
|
38
|
Amitrano F, Krishnan M, Murphy R, Okesene-Gafa KAM, Ji M, Thompson JMD, Taylor RS, Merriman TR, Rush E, McCowan M, McCowan LME, McKinlay CJD. The impact of CREBRF rs373863828 Pacific-variant on infant body composition. Sci Rep 2024; 14:8825. [PMID: 38627436 PMCID: PMC11021527 DOI: 10.1038/s41598-024-59417-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
In Māori and Pacific adults, the CREBRF rs373863828 minor (A) allele is associated with increased body mass index (BMI) but reduced incidence of type-2 and gestational diabetes mellitus. In this prospective cohort study of Māori and Pacific infants, nested within a nutritional intervention trial for pregnant women with obesity and without pregestational diabetes, we investigated whether the rs373863828 A allele is associated with differences in growth and body composition from birth to 12-18 months' corrected age. Infants with and without the variant allele were compared using generalised linear models adjusted for potential confounding by gestation length, sex, ethnicity and parity, and in a secondary analysis, additionally adjusted for gestational diabetes. Carriage of the rs373863828 A allele was not associated with altered growth and body composition from birth to 6 months. At 12-18 months, infants with the rs373863828 A allele had lower whole-body fat mass [FM 1.4 (0.7) vs. 1.7 (0.7) kg, aMD -0.4, 95% CI -0.7, 0.0, P = 0.05; FM index 2.2 (1.1) vs. 2.6 (1.0) kg/m2 aMD -0.6, 95% CI -1.2,0.0, P = 0.04]. However, this association was not significant after adjustment for gestational diabetes, suggesting that it may be mediated, at least in part, by the beneficial effect of CREBRF rs373863828 A allele on maternal glycemic status.
Collapse
Affiliation(s)
| | - Mohanraj Krishnan
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
- Department of Medicine, University of Auckland, Auckland, New Zealand
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rinki Murphy
- Department of Medicine, University of Auckland, Auckland, New Zealand
- Te Whatu Ora, Counties Manukau, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Karaponi A M Okesene-Gafa
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
- Te Whatu Ora, Counties Manukau, Auckland, New Zealand
| | - Maria Ji
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - John M D Thompson
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
- Department of Paediatrics: Child and Youth Health, University of Auckland, Auckland, New Zealand
| | - Rennae S Taylor
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Tony R Merriman
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Elaine Rush
- Faculty of Health and Environmental Science, Auckland University of Technology, Auckland, New Zealand
| | - Megan McCowan
- Te Whatu Ora, Counties Manukau, Auckland, New Zealand
| | - Lesley M E McCowan
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
- Te Whatu Ora, Counties Manukau, Auckland, New Zealand
| | - Christopher J D McKinlay
- Te Whatu Ora, Counties Manukau, Auckland, New Zealand.
- Department of Paediatrics: Child and Youth Health, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
39
|
Imamura M, Maeda S. Perspectives on genetic studies of type 2 diabetes from the genome-wide association studies era to precision medicine. J Diabetes Investig 2024; 15:410-422. [PMID: 38259175 PMCID: PMC10981147 DOI: 10.1111/jdi.14149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/24/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Genome-wide association studies (GWAS) have facilitated a substantial and rapid increase in the number of confirmed genetic susceptibility variants for complex diseases. Approximately 700 variants predisposing individuals to the risk for type 2 diabetes have been identified through GWAS until 2023. From 2018 to 2022, hundreds of type 2 diabetes susceptibility loci with smaller effect sizes were identified through large-scale GWAS with sample sizes of 200,000 to >1 million. The clinical translation of genetic information for type 2 diabetes includes the development of novel therapeutics and risk predictions. Although drug discovery based on loci identified in GWAS remains challenging owing to the difficulty of functional annotation, global efforts have been made to identify novel biological mechanisms and therapeutic targets by applying multi-omics approaches or searching for disease-associated coding variants in isolated founder populations. Polygenic risk scores (PRSs), comprising up to millions of associated variants, can identify individuals with higher disease risk than those in the general population. In populations of European descent, PRSs constructed from base GWAS data with a sample size of approximately 450,000 have predicted the onset of diseases well. However, European GWAS-derived PRSs have limited predictive performance in non-European populations. The predictive accuracy of a PRS largely depends on the sample size of the base GWAS data. The results of GWAS meta-analyses for multi-ethnic groups as base GWAS data and cross-population polygenic prediction methodology have been applied to establish a universal PRS applicable to small isolated ethnic populations.
Collapse
Affiliation(s)
- Minako Imamura
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of MedicineUniversity of the RyukyusNishihara‐ChoJapan
- Division of Clinical Laboratory and Blood TransfusionUniversity of the Ryukyus HospitalNishihara‐ChoJapan
| | - Shiro Maeda
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of MedicineUniversity of the RyukyusNishihara‐ChoJapan
- Division of Clinical Laboratory and Blood TransfusionUniversity of the Ryukyus HospitalNishihara‐ChoJapan
| |
Collapse
|
40
|
Song M, Kwak SH, Kim J. Risk prediction and interaction analysis using polygenic risk score of type 2 diabetes in a Korean population. Sci Rep 2024; 14:6790. [PMID: 38514700 PMCID: PMC10957984 DOI: 10.1038/s41598-024-55945-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 02/29/2024] [Indexed: 03/23/2024] Open
Abstract
Joint modelling of genetic and environmental risk factors can provide important information to predict the risk of type 2 diabetes (T2D). Therefore, to predict the genetic risk of T2D, we constructed a polygenic risk score (PRS) using genotype data of one Korean cohort, KARE (745 cases and 2549 controls), and the genome-wide association study summary statistics of Biobank Japan. We evaluated the performance of PRS in an independent Korean cohort, HEXA (5684 cases and 35,703 controls). Individuals with T2D had a significantly higher mean PRS than controls (0.492 vs. - 0.078, p ≈ 0 ). PRS predicted the risk of T2D with an AUC of 0.658 (95% CI 0.651-0.666). We also evaluated interaction between PRS and waist circumference (WC) in the HEXA cohort. PRS exhibited a significant sub-multiplicative interaction with WC (ORinteraction 0.991, 95% CI 0.987-0.995, pinteraction = 4.93 × 10-6) in T2D. The effect of WC on T2D decreased as PRS increased. The sex-specific analyses produced similar interaction results, revealing a decreased WC effect on T2D as the PRS increased. In conclusion, the risk of WC for T2D may differ depending on PRS and those with a high PRS might develop T2D with a lower WC threshold. Our findings are expected to improve risk prediction for T2D and facilitate the identification of individuals at an increased risk of T2D.
Collapse
Affiliation(s)
- Minsun Song
- Department of Statistics & Research Institute of Natural Sciences, Sookmyung Women's University, Seoul, 04310, Korea
| | - Soo Heon Kwak
- Department of Internal Medicine, Seoul National University Hospital, Seoul, 03080, Korea.
| | - Jihyun Kim
- Department of Statistics, Sookmyung Women's University, Seoul, 04310, Korea
| |
Collapse
|
41
|
Yang W, Yang W, Ma Y, Yan L, Ma X. A novel chromone Schiff base as Zn 2+ turn-on fluorescent chemosensor in a mixed solution. LUMINESCENCE 2024; 39:e4712. [PMID: 38481369 DOI: 10.1002/bio.4712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/04/2024] [Accepted: 02/16/2024] [Indexed: 03/22/2024]
Abstract
In this study, a novel fluorescent chemosensor 1 based on chromone-3-carboxaldehyde Schiff base was synthesized and featured through nuclear magnetic resonance (NMR) and mass spectra. Spectroscopic investigation indicated that the fluorescent sensor showed high selectivity toward Zn2+ over other metal ions and that the detection limit of 1 could reach 10-7 M. These indicated that 1 acted as a highly selective and sensitive fluorescence chemosensor for Zn2+ .
Collapse
Affiliation(s)
- Wensheng Yang
- School of Chemistry and Chemical Engineering, Yulin University, Yulin, China
| | - Wan Yang
- School of Chemistry and Chemical Engineering, Yulin University, Yulin, China
| | - Yajun Ma
- School of Chemistry and Chemical Engineering, Yulin University, Yulin, China
| | - Long Yan
- School of Chemistry and Chemical Engineering, Yulin University, Yulin, China
| | - Xiangrong Ma
- School of Chemistry and Chemical Engineering, Yulin University, Yulin, China
| |
Collapse
|
42
|
Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Barone Gibbs B, Beaton AZ, Boehme AK, Commodore-Mensah Y, Currie ME, Elkind MSV, Evenson KR, Generoso G, Heard DG, Hiremath S, Johansen MC, Kalani R, Kazi DS, Ko D, Liu J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Perman SM, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Tsao CW, Urbut SM, Van Spall HGC, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Palaniappan LP, American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee. 2024 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2024; 149:e347-e913. [PMID: 38264914 PMCID: PMC12146881 DOI: 10.1161/cir.0000000000001209] [Citation(s) in RCA: 845] [Impact Index Per Article: 845.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2024 AHA Statistical Update is the product of a full year's worth of effort in 2023 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. The AHA strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional global data, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
43
|
Kal S, Mahata S, Jati S, Mahata SK. Mitochondrial-derived peptides: Antidiabetic functions and evolutionary perspectives. Peptides 2024; 172:171147. [PMID: 38160808 PMCID: PMC10838678 DOI: 10.1016/j.peptides.2023.171147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
Mitochondrial-derived peptides (MDPs) are a novel class of bioactive microproteins encoded by short open-reading frames (sORF) in mitochondrial DNA (mtDNA). Currently, three types of MDPs have been identified: Humanin (HN), MOTS-c (Mitochondrial ORF within Twelve S rRNA type-c), and SHLP1-6 (small Humanin-like peptide, 1 to 6). The 12 S ribosomal RNA (MT-RNR1) gene harbors the sequence for MOTS-c, whereas HN and SHLP1-6 are encoded by the 16 S ribosomal RNA (MT-RNR2) gene. Special genetic codes are used in mtDNA as compared to nuclear DNA: (i) ATA and ATT are used as start codons in addition to the standard start codon ATG; (ii) AGA and AGG are used as stop codons instead of coding for arginine; (iii) the standard stop codon UGA is used to code for tryptophan. While HN, SHLP6, and MOTS-c are encoded by the H (heavy owing to high guanine + thymine base composition)-strand of the mtDNA, SHLP1-5 are encoded by the L (light owing to less guanine + thymine base composition)-strand. MDPs attenuate disease pathology including Type 1 diabetes (T1D), Type 2 diabetes (T2D), gestational diabetes, Alzheimer's disease (AD), cardiovascular diseases, prostate cancer, and macular degeneration. The current review will focus on the MDP regulation of T2D, T1D, and gestational diabetes along with an emphasis on the evolutionary pressures for conservation of the amino acid sequences of MDPs.
Collapse
Affiliation(s)
- Satadeepa Kal
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sumana Mahata
- Department of Anesthesiology, Riverside University Health System, Moreno Valley, CA, USA
| | - Suborno Jati
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| | - Sushil K Mahata
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
44
|
Lau W, Ali A, Maude H, Andrew T, Swallow DM, Maniatis N. The hazards of genotype imputation when mapping disease susceptibility variants. Genome Biol 2024; 25:7. [PMID: 38172955 PMCID: PMC10763476 DOI: 10.1186/s13059-023-03140-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The cost-free increase in statistical power of using imputation to infer missing genotypes is undoubtedly appealing, but is it hazard-free? This case study of three type-2 diabetes (T2D) loci demonstrates that it is not; it sheds light on why this is so and raises concerns as to the shortcomings of imputation at disease loci, where haplotypes differ between cases and reference panel. RESULTS T2D-associated variants were previously identified using targeted sequencing. We removed these significantly associated SNPs and used neighbouring SNPs to infer them by imputation. We compared imputed with observed genotypes, examined the altered pattern of T2D-SNP association, and investigated the cause of imputation errors by studying haplotype structure. Most T2D variants were incorrectly imputed with a low density of scaffold SNPs, but the majority failed to impute even at high density, despite obtaining high certainty scores. Missing and discordant imputation errors, which were observed disproportionately for the risk alleles, produced monomorphic genotype calls or false-negative associations. We show that haplotypes carrying risk alleles are considerably more common in the T2D cases than the reference panel, for all loci. CONCLUSIONS Imputation is not a panacea for fine mapping, nor for meta-analysing multiple GWAS based on different arrays and different populations. A total of 80% of the SNPs we have tested are not included in array platforms, explaining why these and other such associated variants may previously have been missed. Regardless of the choice of software and reference haplotypes, imputation drives genotype inference towards the reference panel, introducing errors at disease loci.
Collapse
Affiliation(s)
- Winston Lau
- Department of Genetics, Evolution and Environment, UCL Genetics Institute, University College London, London, UK
| | - Aminah Ali
- Department of Genetics, Evolution and Environment, UCL Genetics Institute, University College London, London, UK
| | - Hannah Maude
- Department of Metabolism, Digestion and Reproduction, Section of Genetics and Genomics, London, UK
| | - Toby Andrew
- Department of Metabolism, Digestion and Reproduction, Section of Genetics and Genomics, London, UK
| | - Dallas M Swallow
- Department of Genetics, Evolution and Environment, UCL Genetics Institute, University College London, London, UK
| | - Nikolas Maniatis
- Department of Genetics, Evolution and Environment, UCL Genetics Institute, University College London, London, UK.
| |
Collapse
|
45
|
Chen B, Yu P, Chan WN, Xie F, Zhang Y, Liang L, Leung KT, Lo KW, Yu J, Tse GMK, Kang W, To KF. Cellular zinc metabolism and zinc signaling: from biological functions to diseases and therapeutic targets. Signal Transduct Target Ther 2024; 9:6. [PMID: 38169461 PMCID: PMC10761908 DOI: 10.1038/s41392-023-01679-y] [Citation(s) in RCA: 79] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 01/05/2024] Open
Abstract
Zinc metabolism at the cellular level is critical for many biological processes in the body. A key observation is the disruption of cellular homeostasis, often coinciding with disease progression. As an essential factor in maintaining cellular equilibrium, cellular zinc has been increasingly spotlighted in the context of disease development. Extensive research suggests zinc's involvement in promoting malignancy and invasion in cancer cells, despite its low tissue concentration. This has led to a growing body of literature investigating zinc's cellular metabolism, particularly the functions of zinc transporters and storage mechanisms during cancer progression. Zinc transportation is under the control of two major transporter families: SLC30 (ZnT) for the excretion of zinc and SLC39 (ZIP) for the zinc intake. Additionally, the storage of this essential element is predominantly mediated by metallothioneins (MTs). This review consolidates knowledge on the critical functions of cellular zinc signaling and underscores potential molecular pathways linking zinc metabolism to disease progression, with a special focus on cancer. We also compile a summary of clinical trials involving zinc ions. Given the main localization of zinc transporters at the cell membrane, the potential for targeted therapies, including small molecules and monoclonal antibodies, offers promising avenues for future exploration.
Collapse
Affiliation(s)
- Bonan Chen
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Peiyao Yu
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Wai Nok Chan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Fuda Xie
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Yigan Zhang
- Institute of Biomedical Research, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital and Basic Medical College, Southern Medical University, Guangdong Province Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Kam Tong Leung
- Department of Pediatrics, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwok Wai Lo
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Gary M K Tse
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
- CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
46
|
Al Hageh C, O'Sullivan S, Platt DE, Henschel A, Chacar S, Gauguier D, Abchee A, Alefishat E, Nader M, Zalloua PA. Coronary artery disease patients with rs7904519 (TCF7L2) are at a persistent risk of type 2 diabetes. Diabetes Res Clin Pract 2024; 207:111052. [PMID: 38072013 DOI: 10.1016/j.diabres.2023.111052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/04/2023] [Accepted: 12/07/2023] [Indexed: 02/10/2024]
Abstract
AIMS Type 2 diabetes (T2D) and coronary artery disease (CAD) often coexist and share genetic factors.This study aimed to investigate the common genetic factors underlying T2D and CAD in patients with CAD. METHODS A three-step association approach was conducted: a) a discovery step involving 943 CAD patients with T2D and 1,149 CAD patients without T2D; b) an eliminating step to exclude CAD or T2D specific variants; and c) a replication step using the UK Biobank data. RESULTS Ten genetic loci were associated with T2D in CAD patients. Three variants were specific to either CAD or T2D. Five variants lost significance after adjusting for covariates, while two SNPs remained associated with T2D in CAD patients (rs7904519*G: TCF7L2 and rs17608766*C: GOSR2). The T2D susceptibility rs7904519*G was associated with increased T2D risk, while the CAD susceptibility rs17608766*C was negatively associated with T2D in CAD patients. These associations were replicated in a UK Biobank data, confirming the results. CONCLUSIONS No significant common T2D and CAD susceptibility genetic association was demonstrated indicating distinct disease pathways. However, CAD patients carrying the T2D susceptibility gene TCF7L2 remain at higher risk for developing T2D emphasizing the need for frequent monitoring in this subgroup.
Collapse
Affiliation(s)
- Cynthia Al Hageh
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Siobhan O'Sullivan
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Daniel E Platt
- Computational Biology Center, IBM TJ Watson Research Centre, Yorktown Hgts, NY, USA
| | - Andreas Henschel
- Department of Electrical Engineering and Computer, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Stephanie Chacar
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Dominique Gauguier
- Université Paris Cité, INSERM UMR 1124, 45 rue des Saint-Pères, 75006 Paris, France; McGill University and Genome Quebec Innovation Centre, 740 Doctor Penfield Avenue, Montreal, QC H3A 0G1, Canada
| | | | - Eman Alefishat
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; Department of Pharmacology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi UAE
| | - Moni Nader
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
| | - Pierre A Zalloua
- Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates; Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
47
|
Zhuo C, Zhang Q, Wang L, Ma X, Li R, Ping J, Zhu J, Tian H, Jiang D. Insulin Resistance/Diabetes and Schizophrenia: Potential Shared Genetic Factors and Implications for Better Management of Patients with Schizophrenia. CNS Drugs 2024; 38:33-44. [PMID: 38097908 PMCID: PMC10811033 DOI: 10.1007/s40263-023-01057-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2023] [Indexed: 01/27/2024]
Abstract
Schizophrenia is a complex psychotic disorder with co-occurring conditions, including insulin resistance and type 2 diabetes (T2D). It is well established that T2D and its precursors (i.e., insulin resistance) are more prevalent in patients with schizophrenia who are treated with antipsychotics, as well as in antipsychotic-naïve patients experiencing their first episode of psychosis, compared with the general population. However, the mechanism(s) underlying the increased susceptibility, shared genetics, and possible cause-effect relationship between schizophrenia and T2D remain largely unknown. The objective of this narrative review was to synthesize important studies, including Mendelian randomization (MR) analyses that have integrated genome-wide association studies (GWAS), as well as results from in vitro models, in vivo models, and observational studies of patients with schizophrenia. Both GWAS and MR studies have found that schizophrenia and T2D/insulin resistance share genetic risk factors, and this may mediate a connection between peripheral or brain insulin resistance and T2D with cognition impairment and an increased risk of developing prediabetes and T2D in schizophrenia. Moreover, accumulating evidence supports a causal role for insulin resistance in schizophrenia and emphasizes the importance of a genetic basis for susceptibility to T2D in patients with schizophrenia before they receive psychotic treatment. The present findings and observations may have clinical implications for the development of better strategies to treat patients with schizophrenia, with both pharmacological (i.e., samidorphan, empagliflozin) and/or nonpharmacological (i.e., lifestyle changes) approaches. Additionally, this review may benefit the design of future studies by physicians and clinical investigators.
Collapse
Affiliation(s)
- Chuanjun Zhuo
- Key Laboratory of Sensory Information Processing Abnormalities in Schizophrenia (SIPAS_Lab), Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, No. 1 Zhongshan Road, Hebei District, Tianjin, 300140, China.
- Key Laboratory of Multiple Organs Damage in Patients with Metal Disorder (MODMD_Lab), Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin, 300140, China.
- Laboratory of Psychiatric-Neuroimaging-Genetics and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin, 300222, China.
- GWAS Center of Psychiatry, Wenzhou Seventh Peoples Hospital, Wenzhou, 325000, Zhejiang, China.
| | - Qiuyu Zhang
- Key Laboratory of Sensory Information Processing Abnormalities in Schizophrenia (SIPAS_Lab), Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, No. 1 Zhongshan Road, Hebei District, Tianjin, 300140, China
- Key Laboratory of Multiple Organs Damage in Patients with Metal Disorder (MODMD_Lab), Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin, 300140, China
| | - Lina Wang
- Laboratory of Psychiatric-Neuroimaging-Genetics and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Xiaoyan Ma
- Laboratory of Psychiatric-Neuroimaging-Genetics and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Ranli Li
- Laboratory of Psychiatric-Neuroimaging-Genetics and Co-morbidity (PNGC_Lab), Tianjin Anding Hospital, Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical University, Tianjin, 300222, China
| | - Jing Ping
- GWAS Center of Psychiatry, Wenzhou Seventh Peoples Hospital, Wenzhou, 325000, Zhejiang, China
| | - Jingjing Zhu
- GWAS Center of Psychiatry, Wenzhou Seventh Peoples Hospital, Wenzhou, 325000, Zhejiang, China
| | - Hongjun Tian
- Key Laboratory of Sensory Information Processing Abnormalities in Schizophrenia (SIPAS_Lab), Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, No. 1 Zhongshan Road, Hebei District, Tianjin, 300140, China
- Key Laboratory of Multiple Organs Damage in Patients with Metal Disorder (MODMD_Lab), Tianjin Fourth Center Hospital, Nankai University Affiliated Tianjin Fourth Center Hospital, Tianjin Medical University Affiliated Tianjin Fourth Center Hospital, Tianjin, 300140, China
| | - Deguo Jiang
- GWAS Center of Psychiatry, Wenzhou Seventh Peoples Hospital, Wenzhou, 325000, Zhejiang, China
| |
Collapse
|
48
|
Fan YG, Wu TY, Zhao LX, Jia RJ, Ren H, Hou WJ, Wang ZY. From zinc homeostasis to disease progression: Unveiling the neurodegenerative puzzle. Pharmacol Res 2024; 199:107039. [PMID: 38123108 DOI: 10.1016/j.phrs.2023.107039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/16/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023]
Abstract
Zinc is a crucial trace element in the human body, playing a role in various physiological processes such as oxidative stress, neurotransmission, protein synthesis, and DNA repair. The zinc transporters (ZnTs) family members are responsible for exporting intracellular zinc, while Zrt- and Irt-like proteins (ZIPs) are involved in importing extracellular zinc. These processes are essential for maintaining cellular zinc homeostasis. Imbalances in zinc metabolism have been linked to the development of neurodegenerative diseases. Disruptions in zinc levels can impact the survival and activity of neurons, thereby contributing to the progression of neurodegenerative diseases through mechanisms like cell apoptosis regulation, protein phase separation, ferroptosis, oxidative stress, and neuroinflammation. Therefore, conducting a systematic review of the regulatory network of zinc and investigating the relationship between zinc dysmetabolism and neurodegenerative diseases can enhance our understanding of the pathogenesis of these diseases. Additionally, it may offer new insights and approaches for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yong-Gang Fan
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China.
| | - Ting-Yao Wu
- First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - Ling-Xiao Zhao
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Rong-Jun Jia
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Hang Ren
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Wen-Jia Hou
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhan-You Wang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China.
| |
Collapse
|
49
|
Hung YH, Kim Y, Mitchell SB, Thorn TL, Aydemir TB. Absence of Slc39a14/Zip14 in mouse pancreatic beta cells results in hyperinsulinemia. Am J Physiol Endocrinol Metab 2024; 326:E92-E105. [PMID: 38019082 PMCID: PMC11193513 DOI: 10.1152/ajpendo.00117.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023]
Abstract
Zinc is an essential component of the insulin protein complex synthesized in β cells. The intracellular compartmentalization and distribution of zinc are controlled by 24 transmembrane zinc transporters belonging to the ZnT or Zrt/Irt-like protein (ZIP) family. Downregulation of SLC39A14/ZIP14 has been reported in pancreatic islets of patients with type 2 diabetes (T2D) as well as mouse models of high-fat diet (HFD)- or db/db-induced obesity. Our previous studies observed mild hyperinsulinemia in mice with whole body knockout of Slc39a14 (Zip14 KO). Based on our current secondary data analysis from an integrative single-cell RNA-seq dataset of human whole pancreatic tissue, SLC39A14 (coding ZIP14) is the only other zinc transporter expressed abundantly in human β cells besides well-known zinc transporter SLC30A8 (coding ZnT8). In the present work, using pancreatic β cell-specific knockout of Slc39a14 (β-Zip14 KO), we investigated the role of SLC39A14/ZIP14-mediated intracellular zinc trafficking in glucose-stimulated insulin secretion and subsequent metabolic responses. Glucose-stimulated insulin secretion, zinc concentrations, and cellular localization of ZIP14 were assessed using in vivo, ex vivo, and in vitro assays using β-Zip14 KO, isolated islets, and murine cell line MIN6. Metabolic evaluations were done on both chow- and HFD-fed mice using time-domain nuclear magnetic resonance and a comprehensive laboratory animal monitoring system. ZIP14 localizes on the endoplasmic reticulum regulating intracellular zinc trafficking in β cells and serves as a negative regulator of glucose-stimulated insulin secretion. Deletion of Zip14 resulted in greater glucose-stimulated insulin secretion, increased energy expenditure, and shifted energy metabolism toward fatty acid utilization. HFD caused β-Zip14 KO mice to develop greater islet hyperplasia, compensatory hyperinsulinemia, and mild insulin resistance and hyperglycemia. This study provided new insights into the contribution of metal transporter ZIP14-mediated intracellular zinc trafficking in glucose-stimulated insulin secretion and subsequent metabolic responses.NEW & NOTEWORTHY Metal transporter SLC39A14/ZIP14 is downregulated in pancreatic islets of patients with T2D and mouse models of HFD- or db/db-induced obesity. However, the function of ZIP14-mediated intracellular zinc trafficking in β cells is unknown. Our analyses revealed that SLC39A14 is the only Zn transporter expressed abundantly in human β cells besides SLC30A8. Within the β cells, ZIP14 is localized on the endoplasmic reticulum and serves as a negative regulator of insulin secretion, providing a potential therapeutic target for T2D.
Collapse
Affiliation(s)
- Yu-Han Hung
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
- Department of College of Veterinary Medicine, Cornell University, Ithaca, New York, United States
| | - Yongeun Kim
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Samuel Blake Mitchell
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Trista Lee Thorn
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| | - Tolunay Beker Aydemir
- Division of Nutritional Sciences, Cornell University, Ithaca, New York, United States
| |
Collapse
|
50
|
Al-Romaiyan A, Persaud SJ, Jones PM. Identification of Potential Plant-Derived Pancreatic Beta-Cell-Directed Agents Using New Custom-Designed Screening Method: Gymnema sylvestre as an Example. Molecules 2023; 29:194. [PMID: 38202777 PMCID: PMC10780566 DOI: 10.3390/molecules29010194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/23/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Folk medicines are attractive therapeutic agents for treating type 2 diabetes mellitus (T2DM). Most plant extracts that have been suggested to restore β-cells function were tested in vivo. Some only have been tested in vitro to determine whether they have a direct effect on β-cells islets of Langerhans. Currently, there are no defined criteria for screening of β-cell-directed plant-based remedies as potential antidiabetic agents. SUMMARY In this review, we have identified certain criteria/characteristics that can be used to generate a "screening portfolio" to identify plant extracts as potential β-cell-directed agents for the treatment of T2DM. To validate our screening method, we studied the potential therapeutic efficacy of a Gymnema sylvestre (GS) extract using the screening criteria detailed in the review. Six criteria have been identified and validated using OSA®, a GS extract. By using this screening method, we show that OSA® fulfilled most of the criteria identified for an effective β-cell-directed antidiabetic therapy, being an effective insulin-releasing agent at nontoxic concentrations; maintaining β-cell insulin content by stimulating a concomitant increase in insulin gene transcription; maintaining β-cell mass by protecting against apoptosis; and being effective at maintaining normoglycemia in vivo in a mouse model and a human cohort with T2DM. KEY MESSAGES The present review has highlighted the importance of having a screening portfolio for plant extracts that have potential antidiabetic effects in the treatment of T2DM. We propose that this screening method should be adopted for future studies to identify new β-cell-directed antidiabetic plant derived agents.
Collapse
Affiliation(s)
- Altaf Al-Romaiyan
- Department of Pharmacology and Therapeutics, College of Pharmacy, Kuwait University, Jabriya 046302, Kuwait
| | - Shanta J. Persaud
- Department of Diabetes, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK; (S.J.P.); (P.M.J.)
| | - Peter M. Jones
- Department of Diabetes, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK; (S.J.P.); (P.M.J.)
| |
Collapse
|