1
|
Tans R, Glendorf T, van Herwaarden AE, Venselaar H, van Rijswijck DMH, Wevers RA, Gloerich J, van Gool A, Tack CJ. A rare homozygous INS variant causes adult-onset diabetes. BMJ Open Diabetes Res Care 2024; 12:e004418. [PMID: 39706672 DOI: 10.1136/bmjdrc-2024-004418] [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: 06/22/2024] [Accepted: 11/09/2024] [Indexed: 12/23/2024] Open
Abstract
INTRODUCTION Maturity-onset diabetes of the young (MODY) and neonatal diabetes mellitus (NDM) are the most prevalent causes of monogenic diabetes. MODY is an autosomal dominant condition with onset in childhood and young adulthood, while NDM is defined with diabetes onset within 6 months of age and can be caused by dominant, recessive, X-linked genes or by chromosomal abnormalities. Here, we describe a rare case of monogenic diabetes in a patient who is homozygous for an INS gene variant. RESEARCH DESIGN AND METHODS The index patient, a male diagnosed with type 2 diabetes, was treated with low-dose insulin and metformin. Blood plasma was collected under fasting conditions for analysis. MODY screening was performed using a next-generation sequencing panel. In silico analysis of the insulin variant's three-dimensional structure and its interaction with the insulin receptor was conducted. Insulin receptor affinity and downstream signaling potency were evaluated in vitro. RESULTS Auto-immune diabetes was excluded. A homozygous missense variant of the INS gene (c.130G>A, p.Gly44Arg) was identified in the patient. The combination of three different insulin assays showed that the biosynthesis of proinsulin into insulin was intact. In silico analysis of the mutant insulin 3D structure revealed that the INS variant is likely to affect insulin receptor binding and subsequent in vitro analysis suggested reduced potency in downstream signaling. CONCLUSIONS The homozygous c.130G>A variant in the INS gene results in reduced insulin receptor binding and signaling potency. This, combined with pancreatic β-cell apoptosis or dedifferentiation supposedly, has contributed in the late-onset of monogenic diabetes in the index patient.
Collapse
Affiliation(s)
- Roel Tans
- Translational Metabolic Laboratory, Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| | - Tine Glendorf
- Research and Development, Novo Nordisk A/S, Bagsvaerd, Hovedstaden, Denmark
| | | | - Hanka Venselaar
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Danique M H van Rijswijck
- Translational Metabolic Laboratory, Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ron A Wevers
- Translational Metabolic Laboratory, Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jolein Gloerich
- Translational Metabolic Laboratory, Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| | - Alain van Gool
- Translational Metabolic Laboratory, Department of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Gelderland, Netherlands
| |
Collapse
|
2
|
Horikawa Y, Hosomichi K, Yabe D. Monogenic diabetes. Diabetol Int 2024; 15:679-687. [PMID: 39469542 PMCID: PMC11512936 DOI: 10.1007/s13340-024-00698-6] [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: 01/25/2024] [Indexed: 10/30/2024]
Abstract
Diseases in which genetic factors contribute to nearly 100% of the causation by single-gene mutations are referred to as monogenic disorders or Mendelian genetic diseases. These include neonatal diabetes mellitus (NDM), presenting within the first six months of life, maturity-onset diabetes of the young (MODY), developing later in childhood or adolescence, mitochondrial diabetes (MIDD), and insulin-resistant disorders, etc. On the other hand, common lifestyle-related diseases such as type 2 diabetes (T2DM), hypertension and dyslipidemia are multifactorial, emerging through complex interplay of genetic and environmental factors. The identification of causative genes for diabetes resulting from single-gene abnormalities not only unveils previously unknown mechanisms of insulin secretion and sensitivity at the molecular level but also reveals novel targets for drug development. Moreover, monogenic diabetes in which insulin secretion is impaired serve to clarify the pathophysiology and suggest therapeutic targets for the common multifactorial type 2 diabetes mellitus prevalent in the Japanese population, which is characterized by impaired insulin secretion. In this study, we characterize the various monogenic subtypes of diabetes so far identified.
Collapse
Affiliation(s)
- Yukio Horikawa
- Department of Diabetes, Endocrinology and Metabolism, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, Japan
- Clinical Genetics Center, Gifu University Hospital, Gifu, Japan
- Center for Patient Flow Management, Gifu University Hospital, Gifu, Japan
| | - Kazuyoshi Hosomichi
- Laboratory of Computational Genomics, School of Life Science, Tokyo University of Pharmacy and Life Sciences, Tokyo, 192-0392 Japan
| | - Daisuke Yabe
- Department of Diabetes, Endocrinology and Metabolism, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, Japan
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Osaka, 553-0003 Japan
- Center for One Medicine Innovative Translational Research, Gifu University, Gifu, 501-1194 Japan
- Center for Research, Education and Development for Healthcare Life Design, Gifu University, Gifu, 501-1194 Japan
| |
Collapse
|
3
|
Hasballa I, Maggi D. MODY Only Monogenic? A Narrative Review of the Novel Rare and Low-Penetrant Variants. Int J Mol Sci 2024; 25:8790. [PMID: 39201476 PMCID: PMC11354648 DOI: 10.3390/ijms25168790] [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: 06/03/2024] [Revised: 07/26/2024] [Accepted: 08/12/2024] [Indexed: 09/02/2024] Open
Abstract
Maturity-onset diabetes of the young (MODY) represents the most frequent form of monogenic diabetes mellitus (DM), currently classified in 14 distinct subtypes according to single gene mutations involved in the differentiation and function of pancreatic β-cells. A significant proportion of MODY has unknown etiology, suggesting that the genetic landscape is still to be explored. Recently, novel potentially MODY-causal genes, involved in the differentiation and function of β-cells, have been identified, such as RFX6, NKX2.2, NKX6.1, WFS1, PCBD1, MTOR, TBC1D4, CACNA1E, MNX1, AKT2, NEUROG3, EIF2AK3, GLIS3, HADH, and PTF1A. Genetic and clinical features of MODY variants remain highly heterogeneous, with no direct genotype-phenotype correlation, especially in the low-penetrant subtypes. This is a narrative review of the literature aimed at describing the current state-of-the-art of the novel likely MODY-associated variants. For a deeper understanding of MODY complexity, we also report some related controversies concerning the etiological role of some of the well-known pathological genes and MODY inheritance pattern, as well as the rare association of MODY with autoimmune diabetes. Due to the limited data available, the assessment of MODY-related genes pathogenicity remains challenging, especially in the setting of rare and low-penetrant subtypes. In consideration of the crucial importance of an accurate diagnosis, prognosis and management of MODY, more studies are warranted to further investigate its genetic landscape and the genotype-phenotype correlation, as well as the pathogenetic contribution of the nongenetic modifiers in this cohort of patients.
Collapse
Affiliation(s)
- Iderina Hasballa
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, 16132 Genoa, Italy
| | - Davide Maggi
- Department of Internal Medicine and Medical Specialties (DiMI), University of Genoa, 16132 Genoa, Italy
- Diabetes Clinic, IRCCS Ospedale Policlinico San Martino Genoa, 16132 Genoa, Italy
| |
Collapse
|
4
|
Laugesen K, Gregersen S, Støy J. Novel Approach for Treating Diabetes in a Patient With the Heterozygous Pathogenic Variant R46Q in the Insulin Gene. JCEM CASE REPORTS 2024; 2:luae134. [PMID: 39027635 PMCID: PMC11255476 DOI: 10.1210/jcemcr/luae134] [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: 04/19/2024] [Indexed: 07/20/2024]
Abstract
Maturity-onset diabetes of the young (MODY) is a monogenic disorder of glucose homeostasis with several subtypes, each defined by a distinct genetic etiology. Heterozygous pathogenic variants in the insulin gene are rare causes of MODY, and optimal treatment strategies remain uncertain. Herein we describe a patient with diabetes caused by the heterozygous pathogenic variant R46Q in the insulin gene and the glycemic response to selected antidiabetic treatment regimens. The R46Q pathogenic variant leads to secretion of both mutant and wild-type insulin. In vitro, the mutant insulin is associated with a lower insulin-receptor affinity compared with wild-type insulin and a decline in wild-type insulin secretion. In our patient, treatment with a combination of long- and short-acting insulin led to a decline in hemoglobin A1C (HbA1c), although not to the recommended target. A shift to metformin and subsequent add-on of a sodium-glucose cotransporter 2 inhibitor (SGLT2i) resulted in HbA1c levels of less than 7% (53 mmol/mol) and durable glycemic control. Continuous glucose monitoring and oral glucose tolerance tests confirmed that treatment with metformin and SGLT2i was superior to treatment with insulin. In conclusion, diabetes caused by the pathogenic variant R46Q in the insulin gene may be effectively treated with noninsulin.
Collapse
Affiliation(s)
- Kristina Laugesen
- Department of Clinical Biochemistry, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Søren Gregersen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8200 Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Julie Støy
- Steno Diabetes Center Aarhus, Aarhus University Hospital, 8200 Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
5
|
Kumar KK, Aburawi EH, Ljubisavljevic M, Leow MKS, Feng X, Ansari SA, Emerald BS. Exploring histone deacetylases in type 2 diabetes mellitus: pathophysiological insights and therapeutic avenues. Clin Epigenetics 2024; 16:78. [PMID: 38862980 PMCID: PMC11167878 DOI: 10.1186/s13148-024-01692-0] [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: 02/27/2024] [Accepted: 06/04/2024] [Indexed: 06/13/2024] Open
Abstract
Diabetes mellitus is a chronic disease that impairs metabolism, and its prevalence has reached an epidemic proportion globally. Most people affected are with type 2 diabetes mellitus (T2DM), which is caused by a decline in the numbers or functioning of pancreatic endocrine islet cells, specifically the β-cells that release insulin in sufficient quantity to overcome any insulin resistance of the metabolic tissues. Genetic and epigenetic factors have been implicated as the main contributors to the T2DM. Epigenetic modifiers, histone deacetylases (HDACs), are enzymes that remove acetyl groups from histones and play an important role in a variety of molecular processes, including pancreatic cell destiny, insulin release, insulin production, insulin signalling, and glucose metabolism. HDACs also govern other regulatory processes related to diabetes, such as oxidative stress, inflammation, apoptosis, and fibrosis, revealed by network and functional analysis. This review explains the current understanding of the function of HDACs in diabetic pathophysiology, the inhibitory role of various HDAC inhibitors (HDACi), and their functional importance as biomarkers and possible therapeutic targets for T2DM. While their role in T2DM is still emerging, a better understanding of the role of HDACi may be relevant in improving insulin sensitivity, protecting β-cells and reducing T2DM-associated complications, among others.
Collapse
Affiliation(s)
- Kukkala Kiran Kumar
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 15551, Al Ain, Abu Dhabi, United Arab Emirates
| | - Elhadi Husein Aburawi
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Milos Ljubisavljevic
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Program, Singapore, Singapore
| | - Melvin Khee Shing Leow
- LKC School of Medicine, Nanyang Technological University, Singapore, Singapore
- Dept of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
- Duke-NUS Medical School, Cardiovascular and Metabolic Disorders Program, Singapore, Singapore
| | - Xu Feng
- Department of Biochemistry, YLL School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suraiya Anjum Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates
- ASPIRE Precision Medicine Research Institute, Abu Dhabi, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, PO Box 15551, Al Ain, Abu Dhabi, United Arab Emirates.
- Zayed Center for Health Sciences, United Arab Emirates University, Abu Dhabi, United Arab Emirates.
- ASPIRE Precision Medicine Research Institute, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
6
|
Zou Y, Li S, Chen W, Xu J. Urine-derived stem cell therapy for diabetes mellitus and its complications: progress and challenges. Endocrine 2024; 83:270-284. [PMID: 37801228 DOI: 10.1007/s12020-023-03552-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 09/24/2023] [Indexed: 10/07/2023]
Abstract
Diabetes mellitus (DM) is a chronic and relentlessly progressive metabolic disease characterized by a relative or absolute deficiency of insulin in the body, leading to increased production of advanced glycosylation end products that further enhance oxidative and nitrosative stresses, often leading to multiple macrovascular (cardiovascular disease) and microvascular (e.g., diabetic nephropathy, diabetic retinopathy, and neuropathy) complications, representing the ninth leading cause of death worldwide. Existing medical treatments do not provide a complete cure for DM; thus, stem cell transplantation therapy has become the focus of research on DM and its complications. Urine-derived stem cells (USCs), which are isolated from fresh urine and have biological properties similar to those of mesenchymal stem cells (MSCs), were demonstrated to exert antiapoptotic, antifibrotic, anti-inflammatory, and proangiogenic effects through direct differentiation or paracrine mechanisms and potentially treat patients with DM. USCs also have the advantages of simple noninvasive sample collection procedures, minimal ethical issues, low cost, and easy cell isolation methods and thus have received more attention in regenerative therapies in recent years. This review outlines the biological properties of USCs and the research progress and current limitations of their role in DM and related complications. In summary, USCs have shown good versatility in treating hyperglycemia-impaired target organs in preclinical models, and many challenges remain in translating USC therapies to the clinic.
Collapse
Affiliation(s)
- Yun Zou
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shanshan Li
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wen Chen
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jixiong Xu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
7
|
Cota P, Caliskan ÖS, Bastidas-Ponce A, Jing C, Jaki J, Saber L, Czarnecki O, Taskin D, Blöchinger AK, Kurth T, Sterr M, Burtscher I, Krahmer N, Lickert H, Bakhti M. Insulin regulates human pancreatic endocrine cell differentiation in vitro. Mol Metab 2024; 79:101853. [PMID: 38103636 PMCID: PMC10765254 DOI: 10.1016/j.molmet.2023.101853] [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: 09/14/2023] [Revised: 11/21/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
OBJECTIVE The consequences of mutations in genes associated with monogenic forms of diabetes on human pancreas development cannot be studied in a time-resolved fashion in vivo. More specifically, if recessive mutations in the insulin gene influence human pancreatic endocrine lineage formation is still an unresolved question. METHODS To model the extremely reduced insulin levels in patients with recessive insulin gene mutations, we generated a novel knock-in H2B-Cherry reporter human induced pluripotent stem cell (iPSC) line expressing no insulin upon differentiation to stem cell-derived (SC-) β cells in vitro. Differentiation of iPSCs into the pancreatic and endocrine lineage, combined with immunostaining, Western blotting and proteomics analysis phenotypically characterized the insulin gene deficiency in SC-islets. Furthermore, we leveraged FACS analysis and confocal microscopy to explore the impact of insulin shortage on human endocrine cell induction, composition, differentiation and proliferation. RESULTS Interestingly, insulin-deficient SC-islets exhibited low insulin receptor (IR) signaling when stimulated with glucose but displayed increased IR sensitivity upon treatment with exogenous insulin. Furthermore, insulin shortage did not alter neurogenin-3 (NGN3)-mediated endocrine lineage induction. Nevertheless, lack of insulin skewed the SC-islet cell composition with an increased number in SC-β cell formation at the expense of SC-α cells. Finally, insulin deficiency reduced the rate of SC-β cell proliferation but had no impact on the expansion of SC-α cells. CONCLUSIONS Using iPSC disease modelling, we provide first evidence of insulin function in human pancreatic endocrine lineage formation. These findings help to better understand the phenotypic impact of recessive insulin gene mutations during pancreas development and shed light on insulin gene function beside its physiological role in blood glucose regulation.
Collapse
Affiliation(s)
- Perla Cota
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Özüm Sehnaz Caliskan
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute of Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Aimée Bastidas-Ponce
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Changying Jing
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Munich medical research school (MMRS), Ludwig Maximilian University (LMU), Munich, Germany
| | - Jessica Jaki
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Lama Saber
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Oliver Czarnecki
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Damla Taskin
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
| | - Anna Karolina Blöchinger
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Thomas Kurth
- Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform Core Facility Electron Microscopy and Histology, Technische Universität Dresden, Dresden, Germany
| | - Michael Sterr
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Natalie Krahmer
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute of Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany.
| | - Mostafa Bakhti
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
8
|
de Souza RB, Cabello PH, Rosado EL, Junior MC, de Medeiros Abreu G. What Do We Know about Neonatal Diabetes caused by PDX1 Mutations? Curr Diabetes Rev 2024; 21:e290124226471. [PMID: 38299270 DOI: 10.2174/0115733998265866231204070606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/12/2023] [Accepted: 09/19/2023] [Indexed: 02/02/2024]
Abstract
INTRODUCTION Neonatal diabetes mellitus (NDM) is characterized by severe hyperglycemia, usually diagnosed in the first few months of an individual's life. It is a genetic disease and one of the main forms of monogenic diabetes. Changes in different genes have already been associated with NDM, including changes in the gene PDX1. METHODS In this review, we intend to summarize all neonatal diabetes cases caused by PDX1 mutations reported in the literature. For this purpose, we searched keywords in the literature from PubMed and articles cited by the HGMD database. The search retrieved 84 articles, of which 41 had their full text accessed. After applying the study exclusion criteria, nine articles were included. RESULTS Of those articles, we detected thirteen cases of NDM associated with changes in PDX1; the majority in homozygous or compound heterozygous patients. Until now, variants in the PDX1 gene have been a rare cause of NDM; however, few studies have included the screening of this gene in the investigation of neonatal diabetes. CONCLUSION In this review, we reinforce the importance of the PDX1 gene inclusion in genetic NGS panels for molecular diagnosis of NDM, and systematic morphological and functional exams of the pancreas when NDM is present.
Collapse
Affiliation(s)
- Ritiele Bastos de Souza
- Laboratory of Human Genetics, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Pedro Hernán Cabello
- Laboratory of Human Genetics, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Laboratory of Genetics, School of Health Science, University of Grande Rio, Rio de Janeiro, Brazil
| | - Eliane Lopes Rosado
- Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mário Campos Junior
- Laboratory of Human Genetics, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Gabriella de Medeiros Abreu
- Laboratory of Human Genetics, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Alam A, Dhoundiyal S, Ahmad N, Rao GSNK. Unveiling Diabetes: Categories, Genetics, Diagnostics, Treatments, and Future Horizons. Curr Diabetes Rev 2024; 20:e180823219972. [PMID: 37594107 DOI: 10.2174/1573399820666230818092958] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 06/15/2023] [Accepted: 07/06/2023] [Indexed: 08/19/2023]
Abstract
Diabetes mellitus is a global epidemic affecting millions of individuals worldwide. This comprehensive review aims to provide a thorough understanding of the categorization, disease identity, genetic architecture, diagnosis, and treatment of diabetes. The categorization of diabetes is discussed, with a focus on type 1 and type 2 diabetes, as well as the lesser-known types, type 3 and type 4 diabetes. The geographical variation, age, gender, and ethnic differences in the prevalence of type 1 and type 2 diabetes are explored. The impact of disease identity on disease management and the role of autoimmunity in diabetes are examined. The genetic architecture of diabetes, including the interplay between genotype and phenotype, is discussed to enhance our understanding of the underlying mechanisms. The importance of insulin injection sites and the insulin signalling pathway in diabetes management are highlighted. The diagnostic techniques for diabetes are reviewed, along with advancements for improved differentiation between types. Treatment and management approaches, including medications used in diabetes management are presented. Finally, future perspectives are discussed, emphasizing the need for further research and interventions to address the global burden of diabetes. This review serves as a valuable resource for healthcare professionals, researchers, and policymakers, providing insights to develop targeted strategies for the prevention, diagnosis, and management of this complex disease.
Collapse
Affiliation(s)
- Aftab Alam
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Shivang Dhoundiyal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Niyaz Ahmad
- Department of Pharmaceutical Analysis, Green Research Lab, Green Industrial Company, Second Industrial Area, Riyadh 14334, Saudi Arabia
| | - G S N Koteswara Rao
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| |
Collapse
|
10
|
Yan S, Luo W, Lei L, Zhang Q, Xiu J. Association between serum Klotho concentration and hyperlipidemia in adults: a cross-sectional study from NHANES 2007-2016. Front Endocrinol (Lausanne) 2023; 14:1280873. [PMID: 38027194 PMCID: PMC10668332 DOI: 10.3389/fendo.2023.1280873] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Objective The Klotho protein is a well-documented anti-aging protein known for its diverse biological functions. Hyperlipidemia is an established independent risk factor for various chronic diseases. However, there is limited understanding of the connection between Klotho and hyperlipidemia. The aim was to assess the association between serum Klotho levels and hyperlipidemia among adults. Methods The study included 11,618 individuals from the NHANES database from 2006 to 2017. Hyperlipidemia was diagnosed following the National Cholesterol Education Program guidelines. Serum Klotho concentration was measured by an enzyme-linked immunosorbent assay kit, and the association between Klotho and hyperlipidemia was assessed by a multivariable logistic regression model. Fitted smoothing curves and threshold-effect analysis were employed to describe nonlinear relationships. Results In our multiple logistic regression models, serum Klotho concentration was significantly associated with hyperlipidemia after adjusting for comprehensive confounders (per SD increment odds ratio (OR): 0.91; 95% confidence interval (CI): 0.86-0.97). Compared to individuals in the lowest Klotho quartile, those in the highest quartile exhibited a substantially decreased prevalence of hyperlipidemia (OR: 0.72; 95% CI: 0.58-0.90). Using a two-segment logistic regression model, we identified a U-shaped relationship between serum Klotho concentration and hyperlipidemia, with an inflection point at 1,365.5 pg/mL. Subgroup analysis did not reveal any potential moderating effects. Conclusion This study revealed an inverse relationship between Klotho levels and hyperlipidemia. Further investigation is warranted to explore the underlying mechanism between serum Klotho and hyperlipidemia.
Collapse
Affiliation(s)
| | | | | | - Qiuxia Zhang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiancheng Xiu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
11
|
Goksen D, Evin F, Isik E, Ozen S, Atik T, Ozkinay F, Akcan N, Ozkan B, Buyukinan M, Nuri Ozbek M, Darcan S, Onay H. Molecular diagnosis in patients with monogenic diabetes mellitus, and detection of a novel candidate gene. Diabetes Res Clin Pract 2023; 205:110953. [PMID: 37838154 DOI: 10.1016/j.diabres.2023.110953] [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: 07/14/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/16/2023]
Abstract
AIM We aimed to investigate molecular genetic basis of monogenic diabetes (DM) and novel responsible candidate genes with targeted Next Generation Sequencing (NGS) and Whole Exome Sequencing (WES). METHODS A hundred cases presenting with clinical findings and a family history of monogenic DM were included in the study. Molecular analysis was performed using an NGS panel including 14 genes. Following targeted NGS, WES was planned in cases in whom no variant was detected. RESULTS Thirty different disease-causing variants in seven different genes were detected in thirty-five (35 %) cases with targeted NGS approach. Most common pathogenic variant was found in GCK gene in 25 (25 %) cases. Four different variants were detected in 4 (4 %) patients in ABCC8 gene. In 45 of 65 cases; WES analyses were done. A heterozygous c.2635C > T(p.Gln879Ter) variant was detected in IFIH1 gene in a patient with incidental hyperglycemia. In the segregation analysis affected mother was shown to be heterozygous for the same variant. CONCLUSION Molecular etiology was determined in 35 % cases with the NGS targeted panel. Seventeen novel variants in monogenic DM genes have been identified. A candidate gene determined by WES analysis in a case that could not be diagnosed with NGS panel in this study.
Collapse
Affiliation(s)
- Damla Goksen
- Department of Pediatric Endocrinology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Ferda Evin
- Department of Pediatric Endocrinology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Esra Isik
- Department of Pediatric Genetics, Faculty of Medicine, Ege University, Izmir, Turkey.
| | - Samim Ozen
- Department of Pediatric Endocrinology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Tahir Atik
- Department of Pediatric Genetics, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Ferda Ozkinay
- Department of Pediatric Endocrinology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Nese Akcan
- Department of Pediatric Endocrinology, Faculty of Medicine, Near East University, Nicosia, Cyprus
| | - Behzat Ozkan
- Department of Pediatric Endocrinology, Dr Behçet Uz Çocuk Training and Research Hospital, Izmir, Turkey
| | - Muammer Buyukinan
- Department of Pediatric Endocrinology, Konya Training and Research Hospital, Konya, Turkey
| | - Mehmet Nuri Ozbek
- Department of Pediatric Endocrinology, Mardin Artuklu University, Mardin, Turkey
| | - Sukran Darcan
- Department of Pediatric Endocrinology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Huseyin Onay
- Multigen Genetic Diseases Diagnosis Center, Izmir, Turkey
| |
Collapse
|
12
|
Lee YL, Ting TH, Lim CT, Arrumugam-Arthini C, Karuppiah T, Ling KH. Novel PAX4 variant in a child and family with diabetes mellitus - case report and review of the literature. J Pediatr Endocrinol Metab 2023; 36:988-992. [PMID: 37621150 DOI: 10.1515/jpem-2023-0171] [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: 04/21/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023]
Abstract
OBJECTIVES PAX4 (Paired box 4), a transcription factor crucial in pancreatic beta cell development and function, is a rare cause of maturity-onset diabetes of the young (MODY). What is new? A novel PAX4 variant is verified by family segregation study to be likely pathogenic. A child below 10 years of age diagnosed to have PAX4-MODY, differing from previously reported paediatric cases diagnosed in adolescence. CASE PRESENTATION A child with diabetes diagnosed at age 8 years, harbored a PAX4 variant, c.890G>A (p.Gly297Asp), initially classified as variant of uncertain significance. Eleven family members (7 adults and 4 children) with and without diabetes across 3 generations were genotyped. The variant co-segregated with diabetes or prediabetes across 3 generations of the family. The variant is reclassified as likely pathogenic according to standard guidelines. CONCLUSIONS Genetic testing is essential to confirm PAX4-MODY as the presentation is variable even within the same family. PAX4 mutation needs to be considered in MODY genetic testing in Asian patients.
Collapse
Affiliation(s)
- Yee-Lin Lee
- Department of Paediatrics, Faculty of Medicine and Health Sciences, University Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Tzer-Hwu Ting
- Department of Paediatrics, Faculty of Medicine and Health Sciences, University Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Chong-Teik Lim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Crystal Arrumugam-Arthini
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - Thilakavathy Karuppiah
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Putra Malaysia, UPM Serdang, Selangor, Malaysia
| | - King-Hwa Ling
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Putra Malaysia, UPM Serdang, Selangor, Malaysia
| |
Collapse
|
13
|
Haeri MR. Diabetes and diabesity in the view of proteomics, drug, and plant-derived remedies. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2023; 28:77. [PMID: 38152069 PMCID: PMC10751518 DOI: 10.4103/jrms.jrms_487_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/08/2023] [Accepted: 07/31/2023] [Indexed: 12/29/2023]
Abstract
Diabetes and obesity are highly prevalent in the world. Proteomics is a promising approach to better understanding enzymes, proteins, and signaling molecules involved in diabetes processes which help recognize the basis of the disease better and find suitable new treatments. This study aimed to summarize the molecular mechanisms from the beginning of insulin secretion in response to stimuli to the pathology of the insulin signaling pathway and, finally, the mechanisms of drugs/chemicals remedies that affect this process. The titles and subtitles of this process were determined, and then for each of them, the articles searched in PubMed and ScienceDirect were used. This review article starts the discussion with the molecular basis of insulin biosynthesis, secretion, insulin's mechanism of action, and molecular aspect of diabetes and diabesity (a new term showing the relation between diabetes and obesity) and ends with the drug and plant-derived intervention for hyperglycemia.
Collapse
Affiliation(s)
- Mohammad Reza Haeri
- Department of Clinical Biochemistry, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
- Reference Laboratory, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
14
|
Kõks S. Genomics of Wolfram Syndrome 1 (WFS1). Biomolecules 2023; 13:1346. [PMID: 37759745 PMCID: PMC10527379 DOI: 10.3390/biom13091346] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Wolfram Syndrome (WFS) is a rare, autosomal, recessive neurogenetic disorder that affects many organ systems. It is characterised by diabetes insipidus, diabetes mellites, optic atrophy, and deafness and, therefore, is also known as DIDMOAD. Nearly 15,000-30,000 people are affected by WFS worldwide, and, on average, patients suffering from WFS die at 30 years of age, usually from central respiratory failure caused by massive brain atrophy. The more prevalent of the two kinds of WFS is WFS1, which is a monogenic disease and caused by the loss of the WFS1 gene, whereas WFS2, which is more uncommon, is caused by mutations in the CISD2 gene. Currently, there is no treatment for WFS1 to increase the life expectancy of patients, and the treatments available do not significantly improve their quality of life. Understanding the genetics and the molecular mechanisms of WFS1 is essential to finding a cure. The inability of conventional medications to treat WFS1 points to the need for innovative strategies that must address the fundamental cause: the deletion of the WFS1 gene that leads to the profound ER stress and disturbances in proteostasis. An important approach here is to understand the mechanism of the cell degeneration after the deletion of the WFS1 gene and to describe the differences in these mechanisms for the different tissues. The studies so far have indicated that remarkable clinical heterogeneity is caused by the variable vulnerability caused by WFS1 mutations, and these differences cannot be attributed solely to the positions of mutations in the WFS1 gene. The present review gives a broader overview of the results from genomic studies on the WFS1 mouse model.
Collapse
Affiliation(s)
- Sulev Kõks
- Perron Institute for Neurological and Translational Science, 8 Verdun Street, Nedlands, WA 6009, Australia;
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA 6150, Australia
| |
Collapse
|
15
|
Li M, Popovic N, Wang Y, Chen C, Polychronakos C. Incomplete penetrance and variable expressivity in monogenic diabetes; a challenge but also an opportunity. Rev Endocr Metab Disord 2023; 24:673-684. [PMID: 37165203 DOI: 10.1007/s11154-023-09809-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/01/2023] [Indexed: 05/12/2023]
Abstract
Monogenic Forms of Diabetes (MFD) account for about 3% of all diabetes, and their accurate diagnosis often results in life-changing therapeutic reassignment for the patients. Like other Mendelian diseases, reduced penetrance and variable expressivity are often seen in several different types of MFD, where symptoms develop only in a portion of the persons who carry the pathogenic variant or vary widely in symptom severity and age of onset. This complicates diagnosis and disease management in MFD. In addition to its clinical importance, knowledge of genetic modifiers that confer penetrance and expressivity variability opens possibilities to identify protective genetic variants which may help probe the mechanisms of more common forms of diabetes and shed light in new therapeutic strategies. In this review, we will mainly address penetrance and expressivity variation in different types of MFD, factors that confer such variations and opportunities that come with such knowledge. Related literature was searched in PubMed, Medline and Embase. Papers with publication year from 1974 to 2023 are included. Data are either sourced from literatures or from OMIM, Clinvar and 1000 genome browser.
Collapse
Affiliation(s)
- Meihang Li
- College of pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China.
- Department of Emergency, Department of Endorinology, Maoming People's Hospital, 101 Weimin Road, Maoming, Guangdong, China.
- Montreal Children's Hospital and the Endocrine Genetics Laboratory, Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, China.
- MaiDa Gene Technology, Zhoushan, China.
| | - Natalija Popovic
- Montreal Children's Hospital and the Endocrine Genetics Laboratory, Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, China
| | - Ying Wang
- College of pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, Guangdong, China
| | - Chunbo Chen
- Department of Emergency, Department of Endorinology, Maoming People's Hospital, 101 Weimin Road, Maoming, Guangdong, China
- Department of Critical Care Medicine, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of South University of Science and Technology, Shenzhen, China
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Constantin Polychronakos
- Montreal Children's Hospital and the Endocrine Genetics Laboratory, Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, China
- MaiDa Gene Technology, Zhoushan, China
| |
Collapse
|
16
|
Sun WX, Shu YP, Yang XY, Huang W, Chen J, Yu NN, Zhao M. Effects of folic acid supplementation in pregnant mice on glucose metabolism disorders in male offspring induced by lipopolysaccharide exposure during pregnancy. Sci Rep 2023; 13:7984. [PMID: 37198280 DOI: 10.1038/s41598-023-31690-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 03/15/2023] [Indexed: 05/19/2023] Open
Abstract
The DOHaD theory suggests that adverse environmental factors in early life may lead to the development of metabolic diseases including diabetes and hypertension in adult offspring through epigenetic mechanisms such as DNA methylation. Folic acid (FA) is an important methyl donor in vivo and participates in DNA replication and methylation. The preliminary experimental results of our group demonstrated that lipopolysaccharide (LPS, 50 µg/kg/d) exposure during pregnancy could lead to glucose metabolism disorders in male offspring, but not female offspring; however, the effect of folic acid supplementation on glucose metabolism disorders in male offspring induced by LPS exposure remains unclear. Therefore, in this study, pregnant mice were exposed to LPS on gestational day (GD) 15-17 and were given three doses of FA supplementation (2 mg/kg, 5 mg/kg, or 40 mg/kg) from mating to lactation to explore its effect on glucose metabolism in male offspring and the potential mechanism. This study confirmed that FA supplementation of 5 mg/kg in pregnant mice improved glucose metabolism in LPS-exposed offspring during pregnancy by regulating gene expression.
Collapse
Affiliation(s)
- Wan-Xiao Sun
- Department of Basic Nursing, School of Nursing, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Medical College, Hefei, 230601, Anhui, China
| | - Yi-Ping Shu
- Department of Basic Nursing, School of Nursing, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Xin-Yu Yang
- Department of Basic Nursing, School of Nursing, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Wei Huang
- Department of Basic Nursing, School of Nursing, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Jing Chen
- Department of Basic Nursing, School of Nursing, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Hospital, Hefei, 230022, Anhui, China
| | - Ning-Ning Yu
- Department of Basic Nursing, School of Nursing, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Mei Zhao
- Department of Basic Nursing, School of Nursing, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
17
|
Ataie-Ashtiani S, Forbes B. A Review of the Biosynthesis and Structural Implications of Insulin Gene Mutations Linked to Human Disease. Cells 2023; 12:cells12071008. [PMID: 37048081 PMCID: PMC10093311 DOI: 10.3390/cells12071008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
The discovery of the insulin hormone over 100 years ago, and its subsequent therapeutic application, marked a key landmark in the history of medicine and medical research. The many roles insulin plays in cell metabolism and growth have been revealed by extensive investigations into the structure and function of insulin, the insulin tyrosine kinase receptor (IR), as well as the signalling cascades, which occur upon insulin binding to the IR. In this review, the insulin gene mutations identified as causing disease and the structural implications of these mutations will be discussed. Over 100 studies were evaluated by one reviewing author, and over 70 insulin gene mutations were identified. Mutations may impair insulin gene transcription and translation, preproinsulin trafficking and proinsulin sorting, or insulin-IR interactions. A better understanding of insulin gene mutations and the resultant pathophysiology can give essential insight into the molecular mechanisms underlying impaired insulin biosynthesis and insulin-IR interaction.
Collapse
|
18
|
Abstract
Monogenic diabetes includes several clinical conditions generally characterized by early-onset diabetes, such as neonatal diabetes, maturity-onset diabetes of the young (MODY) and various diabetes-associated syndromes. However, patients with apparent type 2 diabetes mellitus may actually have monogenic diabetes. Indeed, the same monogenic diabetes gene can contribute to different forms of diabetes with early or late onset, depending on the functional impact of the variant, and the same pathogenic variant can produce variable diabetes phenotypes, even in the same family. Monogenic diabetes is mostly caused by impaired function or development of pancreatic islets, with defective insulin secretion in the absence of obesity. The most prevalent form of monogenic diabetes is MODY, which may account for 0.5-5% of patients diagnosed with non-autoimmune diabetes but is probably underdiagnosed owing to insufficient genetic testing. Most patients with neonatal diabetes or MODY have autosomal dominant diabetes. More than 40 subtypes of monogenic diabetes have been identified to date, the most prevalent being deficiencies of GCK and HNF1A. Precision medicine approaches (including specific treatments for hyperglycaemia, monitoring associated extra-pancreatic phenotypes and/or following up clinical trajectories, especially during pregnancy) are available for some forms of monogenic diabetes (including GCK- and HNF1A-diabetes) and increase patients' quality of life. Next-generation sequencing has made genetic diagnosis affordable, enabling effective genomic medicine in monogenic diabetes.
Collapse
|
19
|
Cook TW, Wilstermann AM, Mitchell JT, Arnold NE, Rajasekaran S, Bupp CP, Prokop JW. Understanding Insulin in the Age of Precision Medicine and Big Data: Under-Explored Nature of Genomics. Biomolecules 2023; 13:257. [PMID: 36830626 PMCID: PMC9953665 DOI: 10.3390/biom13020257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Insulin is amongst the human genome's most well-studied genes/proteins due to its connection to metabolic health. Within this article, we review literature and data to build a knowledge base of Insulin (INS) genetics that influence transcription, transcript processing, translation, hormone maturation, secretion, receptor binding, and metabolism while highlighting the future needs of insulin research. The INS gene region has 2076 unique variants from population genetics. Several variants are found near the transcriptional start site, enhancers, and following the INS transcripts that might influence the readthrough fusion transcript INS-IGF2. This INS-IGF2 transcript splice site was confirmed within hundreds of pancreatic RNAseq samples, lacks drift based on human genome sequencing, and has possible elevated expression due to viral regulation within the liver. Moreover, a rare, poorly characterized African population-enriched variant of INS-IGF2 results in a loss of the stop codon. INS transcript UTR variants rs689 and rs3842753, associated with type 1 diabetes, are found in many pancreatic RNAseq datasets with an elevation of the 3'UTR alternatively spliced INS transcript. Finally, by combining literature, evolutionary profiling, and structural biology, we map rare missense variants that influence preproinsulin translation, proinsulin processing, dimer/hexamer secretory storage, receptor activation, and C-peptide detection for quasi-insulin blood measurements.
Collapse
Affiliation(s)
- Taylor W. Cook
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | | | - Jackson T. Mitchell
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Nicholas E. Arnold
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Surender Rajasekaran
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA
| | - Caleb P. Bupp
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Division of Medical Genetics, Corewell Health, Grand Rapids, MI 49503, USA
| | - Jeremy W. Prokop
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
- Office of Research, Corewell Health, Grand Rapids, MI 49503, USA
| |
Collapse
|
20
|
Tian M, Feng Y, Liu Y, Wang H. Case report: A 10-year prognosis of neonatal diabetes caused by a novel INS gene mutation. Front Endocrinol (Lausanne) 2023; 13:1086785. [PMID: 36686471 PMCID: PMC9852905 DOI: 10.3389/fendo.2022.1086785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/20/2022] [Indexed: 01/09/2023] Open
Abstract
Background Neonatal diabetes mellitus (NDM) is a rare form of diabetes. We analyzed a novel insulin gene (INS) mutation of a Chinese permanent neonatal diabetes mellitus (PNDM) patient to explore the clinical and genetic characteristics and put forward some opinions on treatment and its long-term management. Case description A proband was recruited who was diagnosed with permanent neonatal diabetes on his first day after birth. His clinical and follow-up data were collected for 10 years. All of the family members were given an oral glucose tolerance test. Whole exome sequencing was performed on the proband, and the genomic DNA of family members was used for verification by first-generation Sanger sequencing technology. The pathogenic variant was screened according to the American College of Medical Genetics and Genomics classification guidelines and the clinical phenotype of the patient. Diagnostic assessment The proband was diagnosed on the first day after birth, presenting with low birth weight, progressive hyperglycemia, and insulin deficiency. His parents and grandfathers were confirmed to have normal blood sugar levels. A novel homozygous mutation of c.1T>C in the INS gene was detected in the proband, located in the initiation codon. The heterozygous mutations were found in four family members, including his mother, father, and grandfathers. With regular insulin injections, long-term regular follow-up, close monitoring of blood glucose, balanced exercise and diet, and psychological and mutual family support, the blood glucose level was well controlled; there were no acute or chronic complications during this decade. The patient's growth and nervous system development are now no different to those of the same age. Conclusion A favorable prognosis is presented for a permanent neonatal diabetes mellitus (PNDM) patient with a novel mutation in the INS gene in China. The present findings indicate that the genetic diagnosis, early use of insulin, close monitoring of blood glucose, and psychological and mutual family support for patients with INS mutation are necessary for their favorable long-term prognosis.
Collapse
Affiliation(s)
- Mengting Tian
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Disease of Women and Children (Sichuan University) Ministry of Education, Sichuan University, Chengdu, China
| | - Yi Feng
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Disease of Women and Children (Sichuan University) Ministry of Education, Sichuan University, Chengdu, China
| | - Yanyan Liu
- Prenatal Diagnosis Center, Department of Obstetrics & Gynecologic, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hua Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Disease of Women and Children (Sichuan University) Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Dapas M, Dunaif A. Deconstructing a Syndrome: Genomic Insights Into PCOS Causal Mechanisms and Classification. Endocr Rev 2022; 43:927-965. [PMID: 35026001 PMCID: PMC9695127 DOI: 10.1210/endrev/bnac001] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Indexed: 01/16/2023]
Abstract
Polycystic ovary syndrome (PCOS) is among the most common disorders in women of reproductive age, affecting up to 15% worldwide, depending on the diagnostic criteria. PCOS is characterized by a constellation of interrelated reproductive abnormalities, including disordered gonadotropin secretion, increased androgen production, chronic anovulation, and polycystic ovarian morphology. It is frequently associated with insulin resistance and obesity. These reproductive and metabolic derangements cause major morbidities across the lifespan, including anovulatory infertility and type 2 diabetes (T2D). Despite decades of investigative effort, the etiology of PCOS remains unknown. Familial clustering of PCOS cases has indicated a genetic contribution to PCOS. There are rare Mendelian forms of PCOS associated with extreme phenotypes, but PCOS typically follows a non-Mendelian pattern of inheritance consistent with a complex genetic architecture, analogous to T2D and obesity, that reflects the interaction of susceptibility genes and environmental factors. Genomic studies of PCOS have provided important insights into disease pathways and have indicated that current diagnostic criteria do not capture underlying differences in biology associated with different forms of PCOS. We provide a state-of-the-science review of genetic analyses of PCOS, including an overview of genomic methodologies aimed at a general audience of non-geneticists and clinicians. Applications in PCOS will be discussed, including strengths and limitations of each study. The contributions of environmental factors, including developmental origins, will be reviewed. Insights into the pathogenesis and genetic architecture of PCOS will be summarized. Future directions for PCOS genetic studies will be outlined.
Collapse
Affiliation(s)
- Matthew Dapas
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Andrea Dunaif
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
22
|
Yang J, Zhen J, Feng W, Fan Z, Ding L, Yang X, Huang Y, Shu H, Xie J, Li X, Qiao J, Fan Y, Sun J, Li N, Liu T, Wang S, Zhang X, Arvan P, Liu M. IER3IP1 is critical for maintaining glucose homeostasis through regulating the endoplasmic reticulum function and survival of β cells. Proc Natl Acad Sci U S A 2022; 119:e2204443119. [PMID: 36322741 PMCID: PMC9659391 DOI: 10.1073/pnas.2204443119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 10/06/2022] [Indexed: 05/04/2023] Open
Abstract
Recessive mutations in IER3IP1 (immediate early response 3 interacting protein 1) cause a syndrome of microcephaly, epilepsy, and permanent neonatal diabetes (MEDS). IER3IP1 encodes an endoplasmic reticulum (ER) membrane protein, which is crucial for brain development; however, the role of IER3IP1 in β cells remains unknown. We have generated two mouse models with either constitutive or inducible IER3IP1 deletion in β cells, named IER3IP1-βKO and IER3IP1-iβKO, respectively. We found that IER3IP1-βKO causes severe early-onset, insulin-deficient diabetes. Functional studies revealed a markedly dilated β-cell ER along with increased proinsulin misfolding and elevated expression of the ER chaperones, including PDI, ERO1, BiP, and P58IPK. Islet transcriptome analysis confirmed by qRT-PCR revealed decreased expression of genes associated with β-cell maturation, cell cycle, and antiapoptotic genes, accompanied by increased expression of antiproliferation genes. Indeed, multiple independent approaches further demonstrated that IER3IP1-βKO impaired β-cell maturation and proliferation, along with increased condensation of β-cell nuclear chromatin. Inducible β-cell IER3IP1 deletion in adult (8-wk-old) mice induced a similar diabetic phenotype, suggesting that IER3IP1 is also critical for function and survival even after β-cell early development. Importantly, IER3IP1 was decreased in β cells of patients with type 2 diabetes (T2D), suggesting an association of IER3IP1 deficiency with β-cell dysfunction in the more-common form of diabetes. These data not only uncover a critical role of IER3IP1 in β cells but also provide insight into molecular basis of diabetes caused by IER3IP1 mutations.
Collapse
Affiliation(s)
- Jing Yang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jinyang Zhen
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wenli Feng
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhenqian Fan
- Department of Endocrinology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Li Ding
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiaoyun Yang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yumeng Huang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Hua Shu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jing Xie
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xin Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jingting Qiao
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yuxin Fan
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jinhong Sun
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Na Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Tengli Liu
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
- NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300384, China
| | - Shusen Wang
- Organ Transplant Center, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
- NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300384, China
- Human Islet Resource Center, Tianjin First Central Hospital, Tianjin 300384, China
| | - Xiaona Zhang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Peter Arvan
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI 48105
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
23
|
Gruber N, Pinhas-Hamiel O. Diabetes Out-of-the-Box: Diabetes Mellitus and Impairment in Hearing and Vision. Curr Diab Rep 2022; 22:423-432. [PMID: 35789979 DOI: 10.1007/s11892-022-01483-y] [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] [Accepted: 05/18/2022] [Indexed: 01/19/2023]
Abstract
PURPOSEOF REVIEW This review aims to provide an update on the etiologies of diabetes that are due to genetic disorders and that co-occur with impaired hearing or vision and to compare them. The potential mechanisms, including novel treatments, will be detailed. RECENT FINDINGS Wolfram syndrome, Kearns-Sayre syndrome, thiamine-responsive megaloblastic anemia, and maternally inherited diabetes and deafness are genetic disorders characterized by diabetes, impaired hearing, and vision. They differ in mode of inheritance, age at presentation, and the involvement of other organs; they are often misdiagnosed as type 1 or type 2 diabetes. Suspicion of a genetic diabetes syndrome should be raised when pancreatic autoantibodies are negative, other organs are involved, and family history includes diabetes. Correct diagnosis of the various syndromes is important for tailoring the most advanced treatment, preventing disease progression, and enabling proper genetic counseling.
Collapse
Affiliation(s)
- Noah Gruber
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Orit Pinhas-Hamiel
- Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
- The National Juvenile Diabetes Center, Maccabi Health Care Services, Ra'anana, Israel.
| |
Collapse
|
24
|
Zuckerman Levin N, Cohen M, Phillip M, Tenenbaum A, Koren I, Tenenbaum-Rakover Y, Admoni O, Hershkovitz E, Haim A, Mazor Aronovitch K, Zangen D, Strich D, Brener A, Yeshayahu Y, Schon Y, Rachmiel M, Ben-Ari T, Levy-Khademi F, Tibi R, Weiss R, Lebenthal Y, Pinhas-Hamiel O, Shehadeh N. Youth-onset type 2 diabetes in Israel: A national cohort. Pediatr Diabetes 2022; 23:649-659. [PMID: 35521999 DOI: 10.1111/pedi.13351] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/23/2022] [Accepted: 04/21/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Prevalence of youth-onset type 2 diabetes (T2D) has increased worldwide, paralleling the rise in pediatric obesity. Occurrence and clinical manifestations vary regionally and demographically. OBJECTIVES We assessed the incidence, and clinical and demographic manifestations of youth-onset T2D in Israel. METHODS In a national observational study, demographic, clinical, and laboratory data were collected from the medical records of children and adolescents, aged 10-18 years, diagnosed with T2D between the years 2008 and 2019. RESULTS The incidence of youth-onset T2D in Israel increased significantly from 0.63/100,000 in 2008 to 3.41/100,000 in 2019. The study cohort comprised 379 individuals (228 girls [59.7%], 221 Jews [58.3%], mean age 14.7 ± 1.9 years); 73.1% had a positive family history of T2D. Mean body mass index (BMI) z-score was 1.96 ± 0.7, higher in Jews than Arabs. High systolic (≥ 130 mmHg) and diastolic blood pressure (≥ 85 mmHg) were observed in 33.7% and 7.8% of patients, respectively; mean glycosylated hemoglobin (A1c) level at diagnosis was 8.8 ± 2.5%. Dyslipidemia, with high triglyceride (>150 mg/dl) and low HDL-c (<40 mg/dl) levels, was found in 45.6% and 56.5%, respectively. Microalbuminuria and retinopathy were documented at diagnosis, 15.2% and 1.9%, respectively) and increased (36.7% and 4.6%, respectively) at follow-up of 2.9 ± 2.1 years. Criteria of metabolic syndrome were met by 224 (62.2%) patients, and fatty liver documented in 65%, mainly Jews. Psychosocial comorbidity was found in 31%. Treatment with metformin (45.6%), insulin (20.6%), and lifestyle modification (18%) improved glycemic control. CONCLUSION Youth-onset T2D in Israel has increased significantly and presents a unique profile.
Collapse
Affiliation(s)
- Nehama Zuckerman Levin
- Pediatric Diabetes Clinic, Institute of Endocrinology, Diabetes and Metabolism, Rambam Health Care Campus, Haifa, Israel.,The Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Meidan Cohen
- Pediatric Diabetes Clinic, Institute of Endocrinology, Diabetes and Metabolism, Rambam Health Care Campus, Haifa, Israel.,The Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Moshe Phillip
- The Jesse Z and Sara Lea Shafer Institute of Endocrinology and Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ariel Tenenbaum
- The Jesse Z and Sara Lea Shafer Institute of Endocrinology and Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Ilana Koren
- The Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Pediatric Endocrine and Diabetes Unit, Carmel Medical Center, Haifa, Israel
| | - Yardena Tenenbaum-Rakover
- The Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.,Pediatric Endocrine Institute, Ha'Emek Medical Center, Afula, Israel
| | - Osnat Admoni
- Pediatric Endocrine Institute, Ha'Emek Medical Center, Afula, Israel
| | - Eli Hershkovitz
- Pediatric Endocrinology and Diabetes Unit, Soroka Medical Center, Beer Sheva, Israel.,The Faculty of Health Sciences, Goldman School of Medicine, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Alon Haim
- Pediatric Endocrinology and Diabetes Unit, Soroka Medical Center, Beer Sheva, Israel.,The Faculty of Health Sciences, Goldman School of Medicine, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Kineret Mazor Aronovitch
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel.,National Juvenile Diabetes Center, Maccabi Health Care Services, Ra'anana, Israel
| | - David Zangen
- Division of Pediatric Endocrinology, Hadassah Hebrew University Medical Center, Jerusalem, Israel.,Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - David Strich
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,Pediatric Specialist Clinic, Clalit Health Services, Jerusalem, Israel.,Pediatric Endocrinology and Diabetes Unit, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Avivit Brener
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yonatan Yeshayahu
- The Faculty of Health Sciences, Goldman School of Medicine, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Pediatric Endocrine Unit, Department of Pediatrics, Assuta Ashdod Medical Center, Ashdod, Israel
| | - Yossi Schon
- Pediatric Endocrinology Institute, Shamir Medical Center (Assaf Harofeh), Zerifin, Israel
| | - Marianna Rachmiel
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Pediatric Endocrinology Institute, Shamir Medical Center (Assaf Harofeh), Zerifin, Israel
| | - Tal Ben-Ari
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Pediatric Endocrinology and Diabetes Unit, Edith Wolfson Medical Center, Holon, Israel
| | - Floris Levy-Khademi
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,Pediatric Endocrinology and Diabetes Unit, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Rami Tibi
- Pediatric Diabetes Clinic, Institute of Endocrinology, Diabetes and Metabolism, Rambam Health Care Campus, Haifa, Israel.,The Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ram Weiss
- Pediatric Diabetes Clinic, Institute of Endocrinology, Diabetes and Metabolism, Rambam Health Care Campus, Haifa, Israel.,The Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yael Lebenthal
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Orit Pinhas-Hamiel
- Sackler School of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Pediatric Endocrine and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel.,National Juvenile Diabetes Center, Maccabi Health Care Services, Ra'anana, Israel
| | - Naim Shehadeh
- Pediatric Diabetes Clinic, Institute of Endocrinology, Diabetes and Metabolism, Rambam Health Care Campus, Haifa, Israel.,The Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
25
|
O'Sullivan JW, Raghavan S, Marquez-Luna C, Luzum JA, Damrauer SM, Ashley EA, O'Donnell CJ, Willer CJ, Natarajan P. Polygenic Risk Scores for Cardiovascular Disease: A Scientific Statement From the American Heart Association. Circulation 2022; 146:e93-e118. [PMID: 35862132 PMCID: PMC9847481 DOI: 10.1161/cir.0000000000001077] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Cardiovascular disease is the leading contributor to years lost due to disability or premature death among adults. Current efforts focus on risk prediction and risk factor mitigation' which have been recognized for the past half-century. However, despite advances, risk prediction remains imprecise with persistently high rates of incident cardiovascular disease. Genetic characterization has been proposed as an approach to enable earlier and potentially tailored prevention. Rare mendelian pathogenic variants predisposing to cardiometabolic conditions have long been known to contribute to disease risk in some families. However, twin and familial aggregation studies imply that diverse cardiovascular conditions are heritable in the general population. Significant technological and methodological advances since the Human Genome Project are facilitating population-based comprehensive genetic profiling at decreasing costs. Genome-wide association studies from such endeavors continue to elucidate causal mechanisms for cardiovascular diseases. Systematic cataloging for cardiovascular risk alleles also enabled the development of polygenic risk scores. Genetic profiling is becoming widespread in large-scale research, including in health care-associated biobanks, randomized controlled trials, and direct-to-consumer profiling in tens of millions of people. Thus, individuals and their physicians are increasingly presented with polygenic risk scores for cardiovascular conditions in clinical encounters. In this scientific statement, we review the contemporary science, clinical considerations, and future challenges for polygenic risk scores for cardiovascular diseases. We selected 5 cardiometabolic diseases (coronary artery disease, hypercholesterolemia, type 2 diabetes, atrial fibrillation, and venous thromboembolic disease) and response to drug therapy and offer provisional guidance to health care professionals, researchers, policymakers, and patients.
Collapse
|
26
|
Casanova JL, Abel L. From rare disorders of immunity to common determinants of infection: Following the mechanistic thread. Cell 2022; 185:3086-3103. [PMID: 35985287 PMCID: PMC9386946 DOI: 10.1016/j.cell.2022.07.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/11/2022] [Accepted: 07/07/2022] [Indexed: 12/12/2022]
Abstract
The immense interindividual clinical variability during any infection is a long-standing enigma. Inborn errors of IFN-γ and IFN-α/β immunity underlying rare infections with weakly virulent mycobacteria and seasonal influenza virus have inspired studies of two common infections: tuberculosis and COVID-19. A TYK2 genotype impairing IFN-γ production accounts for about 1% of tuberculosis cases, and autoantibodies neutralizing IFN-α/β account for about 15% of critical COVID-19 cases. The discovery of inborn errors and mechanisms underlying rare infections drove the identification of common monogenic or autoimmune determinants of related common infections. This "rare-to-common" genetic and mechanistic approach to infectious diseases may be of heuristic value.
Collapse
Affiliation(s)
- Jean-Laurent Casanova
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France; Department of Pediatrics, Necker Hospital for Sick Children, Paris, France; Howard Hughes Medical Institute, New York, NY, USA.
| | - Laurent Abel
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France; Paris Cité University, Imagine Institute, Paris, France
| |
Collapse
|
27
|
Paz-Pacheco E, Nevado JB, Cutiongco-de la Paz EMC, Jasul GV, Aman AYCL, Ribaya ELA, Francisco MDG, Guanzon MLVV, Uyking-Naranjo ML, Añonuevo-Cruz MCS, Maningat MPDD, Jaring CV, Nacpil-Dominguez PD, Pala-Mohamad AB, Canto AU, Quisumbing JPM, Lat AMM, Bernardo DCC, Mansibang NMM, Calpito KJAC, Ribaya VSD, Ferrer JPY, Biwang JH, Melegrito JB, Deguit CDT, Panerio CEG. Variants of SLC2A10 may be Linked to Poor Response to Metformin. J Endocr Soc 2022; 6:bvac092. [PMID: 35854978 PMCID: PMC9278830 DOI: 10.1210/jendso/bvac092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Indexed: 12/05/2022] Open
Abstract
Purpose A study among Filipinos revealed that only 15% of patients with diabetes achieved glycemic control, and poor response to metformin could be one of the possible reasons. Recent studies demonstrate how genetic variations influence response to metformin. Hence, the present study aimed to determine genetic variants associated with poor response to metformin. Methods Using a candidate variant approach, 195 adult Filipino participants with newly diagnosed type 2 diabetes mellitus (T2DM) were enrolled in a case-control study. Genomic DNA from blood samples were collected. Allelic and genotypic associations of variants with poor response to metformin were determined using exact statistical methods. Results Several polymorphisms were nominally associated with poor response to metformin (Puncorr < 0.05). The most notable is the association of multiple variants in the SLC2A10 gene—rs2425911, rs3092412, and rs2425904—with common additive genetic mode of inheritance. Other variants that have possible associations with poor drug response include rs340874 (PROX-AS1), rs815815 (CALM2), rs1333049 (CDKN2B-AS1), rs2010963 (VEGFA), rs1535435 and rs9494266 (AHI1), rs11128347 (PDZRN3), rs1805081 (NPC1), and rs13266634 (SLC30A8). Conclusion In Filipinos, a trend for the association for several variants was noted, with further observation that several mechanisms may be involved. The results may serve as pilot data for further validation of candidate variants for T2DM pharmacotherapy.
Collapse
Affiliation(s)
- Elizabeth Paz-Pacheco
- Division of Endocrinology, Diabetes and Metabolism, Philippine General Hospital, University of the Philippines Manila, Philippines
| | - Jose B Nevado
- Institutes of Human Genetics, National Institutes of Health, University of the Philippines Manila, Philippines
| | | | - Gabriel V Jasul
- Division of Endocrinology, Diabetes and Metabolism, Philippine General Hospital, University of the Philippines Manila, Philippines
| | | | - Elizabeth Laurize A Ribaya
- Institutes of Human Genetics, National Institutes of Health, University of the Philippines Manila, Philippines
| | - Mark David G Francisco
- Division of Endocrinology, Diabetes and Metabolism, Philippine General Hospital, University of the Philippines Manila, Philippines
| | - Ma Luz Vicenta V Guanzon
- Corazon Locsin Montelibano Memorial Regional Hospital, Bacolod City, Negros Occidental, Philippines
| | | | - Ma Cecille S Añonuevo-Cruz
- Division of Endocrinology, Diabetes and Metabolism, Philippine General Hospital, University of the Philippines Manila, Philippines
| | - Maria Patricia Deanna D Maningat
- Division of Endocrinology, Diabetes and Metabolism, Philippine General Hospital, University of the Philippines Manila, Philippines
| | - Cristina V Jaring
- Division of Endocrinology, Diabetes and Metabolism, Philippine General Hospital, University of the Philippines Manila, Philippines
| | - Paulette D Nacpil-Dominguez
- Division of Endocrinology, Diabetes and Metabolism, Philippine General Hospital, University of the Philippines Manila, Philippines
| | - Aniza B Pala-Mohamad
- Division of Endocrinology, Diabetes and Metabolism, Philippine General Hospital, University of the Philippines Manila, Philippines
| | - Abigail U Canto
- Division of Endocrinology, Diabetes and Metabolism, Philippine General Hospital, University of the Philippines Manila, Philippines
| | - John Paul M Quisumbing
- Division of Endocrinology, Diabetes and Metabolism, Philippine General Hospital, University of the Philippines Manila, Philippines
| | - Annabelle Marie M Lat
- Division of Endocrinology, Diabetes and Metabolism, Philippine General Hospital, University of the Philippines Manila, Philippines
| | - Diane Carla C Bernardo
- Division of Endocrinology, Diabetes and Metabolism, Philippine General Hospital, University of the Philippines Manila, Philippines
| | - Noemie Marie M Mansibang
- Division of Endocrinology, Diabetes and Metabolism, Philippine General Hospital, University of the Philippines Manila, Philippines
| | | | - Vincent Sean D Ribaya
- Institutes of Human Genetics, National Institutes of Health, University of the Philippines Manila, Philippines
| | - Julius Patrick Y Ferrer
- Institutes of Human Genetics, National Institutes of Health, University of the Philippines Manila, Philippines
| | - Jessica H Biwang
- Institutes of Human Genetics, National Institutes of Health, University of the Philippines Manila, Philippines
| | - Jodelyn B Melegrito
- Institutes of Human Genetics, National Institutes of Health, University of the Philippines Manila, Philippines
| | - Christian Deo T Deguit
- Institutes of Human Genetics, National Institutes of Health, University of the Philippines Manila, Philippines
| | - Carlos Emmanuel G Panerio
- Institutes of Human Genetics, National Institutes of Health, University of the Philippines Manila, Philippines
| |
Collapse
|
28
|
Thomas S, Ouhtit A, Al Khatib HA, Eid AH, Mathew S, Nasrallah GK, Emara MM, Al Maslamani MA, Yassine HM. Burden and Disease Pathogenesis of Influenza and Other Respiratory Viruses in Diabetic Patients. J Infect Public Health 2022; 15:412-424. [DOI: 10.1016/j.jiph.2022.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 02/24/2022] [Accepted: 03/07/2022] [Indexed: 02/07/2023] Open
|
29
|
Dhayalan B, Weiss MA. Diabetes-Associated Mutations in Proinsulin Provide a "Molecular Rheostat" of Nascent Foldability. Curr Diab Rep 2022; 22:85-94. [PMID: 35119630 DOI: 10.1007/s11892-022-01447-2] [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] [Accepted: 01/04/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE OF REVIEW Diabetes mellitus (DM) due to toxic misfolding of proinsulin variants provides a monogenic model of endoplasmic reticulum (ER) stress. The mutant proinsulin syndrome (also designated MIDY; Mutant INS-gene-induced Diabetes of Youth or Maturity-onset diabetes of the young 10 (MODY10)) ordinarily presents as permanent neonatal-onset DM, but specific amino-acid substitutions may also present later in childhood or adolescence. This review highlights structural mechanisms of proinsulin folding as inferred from phenotype-genotype relationships. RECENT FINDINGS MIDY mutations most commonly add or remove a cysteine, leading to a variant polypeptide containing an odd number of thiol groups. Such variants are associated with aberrant intermolecular disulfide pairing, ER stress, and neonatal β-cell dysfunction. Non-cysteine-related (NCR) mutations (occurring in both the B and A domains of proinsulin) define distinct determinants of foldability and vary in severity. The range of ages of onset, therefore, reflects a "molecular rheostat" connecting protein biophysics to quality-control ER checkpoints. Because in most mammalian cell lines even wild-type proinsulin exhibits limited folding efficiency, molecular barriers to folding uncovered by NCR MIDY mutations may pertain to β-cell dysfunction in non-syndromic type 2 DM due to INS-gene overexpression in the face of peripheral insulin resistance. Recent studies of MIDY mutations and related NCR variants, combining molecular and cell-based approaches, suggest that proinsulin has evolved at the edge of non-foldability. Chemical protein synthesis promises to enable comparative studies of "non-foldable" proinsulin variants to define key steps in wild-type biosynthesis. Such studies may create opportunities for novel therapeutic approaches to non-syndromic type 2 DM.
Collapse
Affiliation(s)
- Balamurugan Dhayalan
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Michael A Weiss
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Chemistry, Indiana University, Bloomington, IN, 47405, USA.
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
30
|
Zhang F, Li Q, Wu J, Ruan H, Sun C, Zhu J, Song Q, Wei X, Shi Y, Zhu L. Total Flavonoids of Drynariae Rhizoma Improve Glucocorticoid-Induced Osteoporosis of Rats: UHPLC-MS-Based Qualitative Analysis, Network Pharmacology Strategy and Pharmacodynamic Validation. Front Endocrinol (Lausanne) 2022; 13:920931. [PMID: 35846330 PMCID: PMC9279576 DOI: 10.3389/fendo.2022.920931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Glucocorticoid-induced osteoporosis (GIOP) is a common form of secondary osteoporosis caused by the protracted or a large dosage of glucocorticoids (GCs). Total flavonoids of Drynariae rhizoma (TFDR) have been widely used in treating postmenopausal osteoporosis (POP). However, their therapeutic effects and potential mechanism against GIOP have not been fully elucidated. METHODS Ultra-high-performance liquid chromatography coupled with electrospray ionization quadrupole time-of-flight mass spectrometry (UHPLC-ESIQ-TOF-MS) experiments were performed for qualitative analysis. We performed hematoxylin-eosin (HE) staining and microcomputed tomography (micro-CT) analysis to detect the changes in bone microstructure. The changes in biochemical parameters in the serum samples were determined by performing an enzyme-linked immunosorbent assay (ELISA). The prediction results of network pharmacology were verified via quantitative real-time polymerase chain reaction (qRT-PCR) to elucidate the potential mechanism of TFDR against GIOP. RESULTS A total of 191 ingredients were identified in vitro and 48 ingredients in vivo. In the in-vivo experiment, the levels of the serum total cholesterol (TC), the serum triglyceride (TG), Leptin (LEP), osteocalcin (OC), osteoprotegerin (OPG), bone morphogenetic protein-2 (BMP-2), propeptide of type I procollagen (PINP), tartrate-resistant acid phosphatase (TRACP) and type-I collagen carboxy-terminal peptide (CTX-1) in the TFDR group significantly changed compared with those in the GIOP group. Moreover, the TFDR group showed an improvement in bone mineral density and bone microstructure. Based on the results of network pharmacology analysis, 67 core targets were selected to construct the network and perform PPI analysis as well as biological enrichment analysis. Five of the targets with high "degree value" had differential gene expression between groups using qRT-PCR. CONCLUSION TFDR, which may play a crucial role between adipose metabolism and bone metabolism, may be a novel remedy for the prevention and clinical treatment of GIOP.
Collapse
Affiliation(s)
- Fangqing Zhang
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiuyue Li
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiashuo Wu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haonan Ruan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chuanrui Sun
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jia Zhu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qinghui Song
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xu Wei
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Xu Wei, ; Yue Shi, ; Liguo Zhu,
| | - Yue Shi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Xu Wei, ; Yue Shi, ; Liguo Zhu,
| | - Liguo Zhu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Xu Wei, ; Yue Shi, ; Liguo Zhu,
| |
Collapse
|
31
|
Silverstein A, Dudaev A, Studneva M, Aitken J, Blokh S, Miller AD, Tanasova S, Rose N, Ryals J, Borchers C, Nordstrom A, Moiseyakh M, Herrera AS, Skomorohov N, Marshall T, Wu A, Cheng RH, Syzko K, Cotter PD, Podzyuban M, Thilly W, Smith PD, Barach P, Bouri K, Schoenfeld Y, Matsuura E, Medvedeva V, Shmulevich I, Cheng L, Seegers P, Khotskaya Y, Flaherty K, Dooley S, Sorenson EJ, Ross M, Suchkov S. Evolution of biomarker research in autoimmunity conditions for health professionals and clinical practice. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 190:219-276. [DOI: 10.1016/bs.pmbts.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
32
|
Clark KC, Kwitek AE. Multi-Omic Approaches to Identify Genetic Factors in Metabolic Syndrome. Compr Physiol 2021; 12:3045-3084. [PMID: 34964118 PMCID: PMC9373910 DOI: 10.1002/cphy.c210010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Metabolic syndrome (MetS) is a highly heritable disease and a major public health burden worldwide. MetS diagnosis criteria are met by the simultaneous presence of any three of the following: high triglycerides, low HDL/high LDL cholesterol, insulin resistance, hypertension, and central obesity. These diseases act synergistically in people suffering from MetS and dramatically increase risk of morbidity and mortality due to stroke and cardiovascular disease, as well as certain cancers. Each of these component features is itself a complex disease, as is MetS. As a genetically complex disease, genetic risk factors for MetS are numerous, but not very powerful individually, often requiring specific environmental stressors for the disease to manifest. When taken together, all sequence variants that contribute to MetS disease risk explain only a fraction of the heritable variance, suggesting additional, novel loci have yet to be discovered. In this article, we will give a brief overview on the genetic concepts needed to interpret genome-wide association studies (GWAS) and quantitative trait locus (QTL) data, summarize the state of the field of MetS physiological genomics, and to introduce tools and resources that can be used by the physiologist to integrate genomics into their own research on MetS and any of its component features. There is a wealth of phenotypic and molecular data in animal models and humans that can be leveraged as outlined in this article. Integrating these multi-omic QTL data for complex diseases such as MetS provides a means to unravel the pathways and mechanisms leading to complex disease and promise for novel treatments. © 2022 American Physiological Society. Compr Physiol 12:1-40, 2022.
Collapse
Affiliation(s)
- Karen C Clark
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Anne E Kwitek
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
33
|
Redondo MJ, Balasubramanyam A. Toward an Improved Classification of Type 2 Diabetes: Lessons From Research into the Heterogeneity of a Complex Disease. J Clin Endocrinol Metab 2021; 106:e4822-e4833. [PMID: 34291809 PMCID: PMC8787852 DOI: 10.1210/clinem/dgab545] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Accumulating evidence indicates that type 2 diabetes (T2D) is phenotypically heterogeneous. Defining and classifying variant forms of T2D are priorities to better understand its pathophysiology and usher clinical practice into an era of "precision diabetes." EVIDENCE ACQUISITION AND METHODS We reviewed literature related to heterogeneity of T2D over the past 5 decades and identified a range of phenotypic variants of T2D. Their descriptions expose inadequacies in current classification systems. We attempt to link phenotypically diverse forms to pathophysiology, explore investigative methods that have characterized "atypical" forms of T2D on an etiological basis, and review conceptual frameworks for an improved taxonomy. Finally, we propose future directions to achieve the goal of an etiological classification of T2D. EVIDENCE SYNTHESIS Differences among ethnic and racial groups were early observations of phenotypic heterogeneity. Investigations that uncover complex interactions of pathophysiologic pathways leading to T2D are supported by epidemiological and clinical differences between the sexes and between adult and youth-onset T2D. Approaches to an etiological classification are illustrated by investigations of atypical forms of T2D, such as monogenic diabetes and syndromes of ketosis-prone diabetes. Conceptual frameworks that accommodate heterogeneity in T2D include an overlap between known diabetes types, a "palette" model integrated with a "threshold hypothesis," and a spectrum model of atypical diabetes. CONCLUSION The heterogeneity of T2D demands an improved, etiological classification scheme. Excellent phenotypic descriptions of emerging syndromes in different populations, continued clinical and molecular investigations of atypical forms of diabetes, and useful conceptual models can be utilized to achieve this important goal.
Collapse
Affiliation(s)
- Maria J Redondo
- Section of Diabetes and Endocrinology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital, Houston, TX 77030, USA
| | - Ashok Balasubramanyam
- Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
34
|
Heller S, Li Z, Lin Q, Geusz R, Breunig M, Hohwieler M, Zhang X, Nair GG, Seufferlein T, Hebrok M, Sander M, Julier C, Kleger A, Costa IG. Transcriptional changes and the role of ONECUT1 in hPSC pancreatic differentiation. Commun Biol 2021; 4:1298. [PMID: 34789845 PMCID: PMC8599846 DOI: 10.1038/s42003-021-02818-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/24/2021] [Indexed: 02/07/2023] Open
Abstract
Cell type specification during pancreatic development is tightly controlled by a transcriptional and epigenetic network. The precise role of most transcription factors, however, has been only described in mice. To convey such concepts to human pancreatic development, alternative model systems such as pancreatic in vitro differentiation of human pluripotent stem cells can be employed. Here, we analyzed stage-specific RNA-, ChIP-, and ATAC-sequencing data to dissect transcriptional and regulatory mechanisms during pancreatic development. Transcriptome and open chromatin maps of pancreatic differentiation from human pluripotent stem cells provide a stage-specific pattern of known pancreatic transcription factors and indicate ONECUT1 as a crucial fate regulator in pancreas progenitors. Moreover, our data suggest that ONECUT1 is also involved in preparing pancreatic progenitors for later endocrine specification. The dissection of the transcriptional and regulatory circuitry revealed an important role for ONECUT1 within such network and will serve as resource to study human development and disease.
Collapse
Affiliation(s)
- Sandra Heller
- grid.410712.1Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Zhijian Li
- grid.1957.a0000 0001 0728 696XInstitute for Computational Genomics, RWTH Aachen University Medical School, Aachen, Germany
| | - Qiong Lin
- grid.420044.60000 0004 0374 4101Bayer AG, Research & Development, Pharmaceuticals, Bioinformatics, Berlin, Germany
| | - Ryan Geusz
- grid.266100.30000 0001 2107 4242Pediatric Diabetes Research Center (PDRC) at the University of California, San Diego, USA
| | - Markus Breunig
- grid.410712.1Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Meike Hohwieler
- grid.410712.1Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Xi Zhang
- grid.410712.1Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Gopika G. Nair
- grid.266102.10000 0001 2297 6811Diabetes Center at the University of California, San Francisco, USA
| | - Thomas Seufferlein
- grid.410712.1Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Matthias Hebrok
- grid.266102.10000 0001 2297 6811Diabetes Center at the University of California, San Francisco, USA
| | - Maike Sander
- grid.266100.30000 0001 2107 4242Pediatric Diabetes Research Center (PDRC) at the University of California, San Diego, USA
| | - Cécile Julier
- grid.4444.00000 0001 2112 9282Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR-8104, Paris, France
| | - Alexander Kleger
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany.
| | - Ivan G. Costa
- grid.1957.a0000 0001 0728 696XInstitute for Computational Genomics, RWTH Aachen University Medical School, Aachen, Germany
| |
Collapse
|
35
|
Mutations and variants of ONECUT1 in diabetes. Nat Med 2021; 27:1928-1940. [PMID: 34663987 DOI: 10.1038/s41591-021-01502-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/13/2021] [Indexed: 12/12/2022]
Abstract
Genes involved in distinct diabetes types suggest shared disease mechanisms. Here we show that One Cut Homeobox 1 (ONECUT1) mutations cause monogenic recessive syndromic diabetes in two unrelated patients, characterized by intrauterine growth retardation, pancreas hypoplasia and gallbladder agenesis/hypoplasia, and early-onset diabetes in heterozygous relatives. Heterozygous carriers of rare coding variants of ONECUT1 define a distinctive subgroup of diabetic patients with early-onset, nonautoimmune diabetes, who respond well to diabetes treatment. In addition, common regulatory ONECUT1 variants are associated with multifactorial type 2 diabetes. Directed differentiation of human pluripotent stem cells revealed that loss of ONECUT1 impairs pancreatic progenitor formation and a subsequent endocrine program. Loss of ONECUT1 altered transcription factor binding and enhancer activity and NKX2.2/NKX6.1 expression in pancreatic progenitor cells. Collectively, we demonstrate that ONECUT1 controls a transcriptional and epigenetic machinery regulating endocrine development, involved in a spectrum of diabetes, encompassing monogenic (recessive and dominant) as well as multifactorial inheritance. Our findings highlight the broad contribution of ONECUT1 in diabetes pathogenesis, marking an important step toward precision diabetes medicine.
Collapse
|
36
|
Li T, Quan H, Zhang H, Lin L, Ou Q, Chen K. Silencing cyclophilin A improves insulin secretion, reduces cell apoptosis, and alleviates inflammation as well as oxidant stress in high glucose-induced pancreatic β-cells via MAPK/NF-kb signaling pathway. Bioengineered 2021; 11:1047-1057. [PMID: 32970961 PMCID: PMC8291783 DOI: 10.1080/21655979.2020.1823729] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cyclophilin A is increased in the plasm of diabetic patients, while its effects on high glucose (HG)-stimulated pancreatic β-cells are still pending. The aim of this research is to investigate the effects of cyclophilin A inhibition on HG-challenged pancreatic β-cells. For investigating the effects of cyclophilin A decrease on HG-induced pancreatic β-cells, the cells were separated into normal glucose (NG), Mannitol, HG, HG + shRNA-NC, and HG + shRNA-Cyclophilin A-1 groups. The protein and mRNA expression were detected via Western blot and qRT-PCR. CCK-8 assay and flow cytometry were employed for assessing cell viability and apoptosis. The levels of oxidative stress, inflammation, and insulin secretion were detected by corresponding kits. The cyclophilin A was higher in HG group. Knockdown of cyclophilin A was able to increase insulin secretion, decrease cell apoptosis, and alleviate inflammation as well as oxidant stress in HG-treated pancreatic β-cells via MAPK/NF-kb pathway. Taken together, Cyclophilin A, highly expressed in pancreatic β-cells induced by HG, is a promising therapeutic target for diabetes. Knockdown of cyclophilin A has protective effects against HG-challenged pancreatic β-cells via regulation of MAPK/NF-kb pathway. The findings in this study provided a new strategy for diabetic treatment and paved the way for future researches on diabetes treatment.
Collapse
Affiliation(s)
- Tangying Li
- Department of Health Care Centre, Hainan General Hospital , Haikou, Hainan, China
| | - Huibiao Quan
- Department of Endocrinology, Hainan General Hospital , Haikou, Hainan, China
| | - Huachuan Zhang
- Department of Endocrinology Laboratory, Hainan General Hospital , Haikou, Hainan, China
| | - Leweihua Lin
- Department of Endocrinology, Hainan General Hospital , Haikou, Hainan, China
| | - Qianying Ou
- Department of Endocrinology, Hainan General Hospital , Haikou, Hainan, China
| | - Kaining Chen
- Department of Endocrinology, Hainan General Hospital , Haikou, Hainan, China
| |
Collapse
|
37
|
Lei L, Bai YH, Jiang HY, He T, Li M, Wang JP. A bioinformatics analysis of the contribution of m6A methylation to the occurrence of diabetes mellitus. Endocr Connect 2021; 10:1253-1265. [PMID: 34486983 PMCID: PMC8558884 DOI: 10.1530/ec-21-0328] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022]
Abstract
N6-methyladenosine (m6A) methylation has been reported to play a role in type 2 diabetes (T2D). However, the key component of m6A methylation has not been well explored in T2D. This study investigates the biological role and the underlying mechanism of m6A methylation genes in T2D. The Gene Expression Omnibus (GEO) database combined with the m6A methylation and transcriptome data of T2D patients were used to identify m6A methylation differentially expressed genes (mMDEGs). Ingenuity pathway analysis (IPA) was used to predict T2D-related differentially expressed genes (DEGs). Gene ontology (GO) term enrichment and the Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to determine the biological functions of mMDEGs. Gene set enrichment analysis (GSEA) was performed to further confirm the functional enrichment of mMDEGs and determine candidate hub genes. The least absolute shrinkage and selection operator (LASSO) regression analysis was carried out to screen for the best predictors of T2D, and RT-PCR and Western blot were used to verify the expression of the predictors. A total of 194 overlapping mMDEGs were detected. GO, KEGG, and GSEA analysis showed that mMDEGs were enriched in T2D and insulin signaling pathways, where the insulin gene (INS), the type 2 membranal glycoprotein gene (MAFA), and hexokinase 2 (HK2) gene were found. The LASSO regression analysis of candidate hub genes showed that the INS gene could be invoked as a predictive hub gene for T2D. INS, MAFA,and HK2 genes participate in the T2D disease process, but INS can better predict the occurrence of T2D.
Collapse
Affiliation(s)
- Lei Lei
- Department of Nephrology, The Second Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Yi-Hua Bai
- Department of Nephrology, The Second Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
- Correspondence should be addressed to Y-H Bai:
| | - Hong-Ying Jiang
- Department of Nephrology, The Second Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Ting He
- Department of Nephrology, The Second Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Meng Li
- Department of Nephrology, The Second Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| | - Jia-Ping Wang
- Department of Radiology, The Second Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
38
|
Yahaya TO, Anyebe DA. Genes predisposing to neonatal diabetes mellitus and pathophysiology: Current findings. J Neonatal Perinatal Med 2021; 13:543-553. [PMID: 32333556 DOI: 10.3233/npm-190353] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Precision medicine, described as a therapeutic procedure in which complex diseases are treated based on the causal gene and pathophysiology, is being considered for diabetes mellitus (DM). To this end, several monogenetic mutations in the beta cells have been linked with neonatal diabetes mellitus (NDM), however, the list of suspect genes is expansive, necessitating an update. This study, therefore, provides an update on NDM candidate genes and pathophysiology. RESULTS Reputable online academic databases were searched for relevant information, which led to the identification of 43 genes whose mutations are linked to the condition. Of the linked genes, mutations in the KCNJ11, ABCC8, and INS genes as well as the genes on 6q24 chromosomal region are the most frequently implicated. Mutations in these genes can cause pancreatic agenesis and developmental errors, resulting in NDM in the first six to twelve months of birth. The clinical presentations of NDM include frequent urination, rapid breathing, and dehydration, among others. CONCLUSIONS Monogenetic mutations in the beta cells may cause NDM with distinct pathophysiology from other DM. Treatment options that target NDM candidate genes and pathophysiology may lead to an improved treatment compared with the present generalized treatment for all forms of DM.
Collapse
Affiliation(s)
- T O Yahaya
- Department of Biology, Federal University Birnin Kebbi, Nigeria
| | - D A Anyebe
- Department of Biochemistry and Molecular Biology, Federal University Birnin Kebbi, Nigeria
| |
Collapse
|
39
|
Ali Khan I. Do second generation sequencing techniques identify documented genetic markers for neonatal diabetes mellitus? Heliyon 2021; 7:e07903. [PMID: 34584998 PMCID: PMC8455689 DOI: 10.1016/j.heliyon.2021.e07903] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 01/15/2021] [Accepted: 08/27/2021] [Indexed: 12/24/2022] Open
Abstract
Neonatal diabetes mellitus (NDM) is noted as a genetic, heterogeneous, and rare disease in infants. NDM occurs due to a single-gene mutation in neonates. A common source for developing NDM in an infant is the existence of mutations/variants in the KCNJ11 and ABCC8 genes, encoding the subunits of the voltage-dependent potassium channel. Both KCNJ11 and ABCC8 genes are useful in diagnosing monogenic diabetes during infancy. Genetic analysis was previously performed using first-generation sequencing techniques, such as DNA-Sanger sequencing, which uses chain-terminating inhibitors. Sanger sequencing has certain limitations; it can screen a limited region of exons in one gene, but it cannot screen large regions of the human genome. In the last decade, first generation sequencing techniques have been replaced with second-generation sequencing techniques, such as next-generation sequencing (NGS), which sequences nucleic-acids more rapidly and economically than Sanger sequencing. NGS applications are involved in whole exome sequencing (WES), whole genome sequencing (WGS), and targeted gene panels. WES characterizes a substantial breakthrough in human genetics. Genetic testing for custom genes allows the screening of the complete gene, including introns and exons. The aim of this review was to confirm if the 22 genetic variations previously documented to cause NDM by Sanger sequencing could be detected using second generation sequencing techniques. The author has cross-checked global studies performed in NDM using NGS, ES/WES, WGS, and targeted gene panels as second-generation sequencing techniques; WES confirmed the similar variants, which have been previously documented with Sanger sequencing. WES is documented as a powerful tool and WGS as the most comprehensive test for verified the documented variants, as well as novel enhancers. This review recommends for the future studies should be performed with second generation sequencing techniques to identify the verified 22 genetic and novel variants by screening in NDM (PNDM or TNMD) children.
Collapse
Affiliation(s)
- Imran Ali Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, PO Box-10219, Riyadh, 11433, Saudi Arabia
| |
Collapse
|
40
|
Shi D, Motamed M, Mejía-Benítez A, Li L, Lin E, Budhram D, Kaur Y, Meyre D. Genetic syndromes with diabetes: A systematic review. Obes Rev 2021; 22:e13303. [PMID: 34268868 DOI: 10.1111/obr.13303] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 01/19/2023]
Abstract
Previous reviews and clinical guidelines have identified 10-20 genetic syndromes associated with diabetes, but no systematic review has been conducted to date. We provide the first comprehensive catalog for syndromes with diabetes mellitus. We conducted a systematic review of MEDLINE, Embase, CENTRAL, PubMed, OMIM, and Orphanet databases for case reports, case series, and observational studies published between 1946 and January 15, 2020, that described diabetes mellitus in adults and children with monogenic or chromosomal syndromes. Our literature search identified 7,122 studies, of which 160 fulfilled inclusion criteria. Our analysis of these studies found 69 distinct diabetes syndromes. Thirty (43.5%) syndromes included diabetes mellitus as a cardinal clinical feature, and 56 (81.2%) were fully genetically elucidated. Sixty-three syndromes (91.3%) were described more than once in independent case reports, of which 59 (93.7%) demonstrated clinical heterogeneity. Syndromes associated with diabetes mellitus are more numerous and diverse than previously anticipated. While knowledge of the syndromes is limited by their low prevalence, future reviews will be needed as more cases are identified. The genetic etiologies of these syndromes are well elucidated and provide potential avenues for future gene identification efforts, aid in diagnosis and management, gene therapy research, and developing personalized medicine treatments.
Collapse
Affiliation(s)
- Daniel Shi
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada.,Faculty of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Mehras Motamed
- Faculty of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Aurora Mejía-Benítez
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Leon Li
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Ethan Lin
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Dalton Budhram
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada.,Faculty of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Yuvreet Kaur
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada.,Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - David Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, and Nutrition, University Hospital of Nancy, Nancy, France.,Faculty of Medicine of Nancy INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy, France
| |
Collapse
|
41
|
Polymorphisms in GLIS3 and susceptibility to diabetes mellitus: A systematic review and meta-analysis. Meta Gene 2021. [DOI: 10.1016/j.mgene.2021.100898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
42
|
Ikegami H, Babaya N, Noso S. β-Cell failure in diabetes: Common susceptibility and mechanisms shared between type 1 and type 2 diabetes. J Diabetes Investig 2021; 12:1526-1539. [PMID: 33993642 PMCID: PMC8409822 DOI: 10.1111/jdi.13576] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022] Open
Abstract
Diabetes mellitus is etiologically classified into type 1, type 2 and other types of diabetes. Despite distinct etiologies and pathogenesis of these subtypes, many studies have suggested the presence of shared susceptibilities and underlying mechanisms in β-cell failure among different types of diabetes. Understanding these susceptibilities and mechanisms can help in the development of therapeutic strategies regardless of the diabetes subtype. In this review, we discuss recent evidence indicating the shared genetic susceptibilities and common molecular mechanisms between type 1, type 2 and other types of diabetes, and highlight the future prospects as well.
Collapse
Affiliation(s)
- Hiroshi Ikegami
- Department of Endocrinology, Metabolism and DiabetesFaculty of MedicineKindai UniversityOsaka‐sayama, OsakaJapan
| | - Naru Babaya
- Department of Endocrinology, Metabolism and DiabetesFaculty of MedicineKindai UniversityOsaka‐sayama, OsakaJapan
| | - Shinsuke Noso
- Department of Endocrinology, Metabolism and DiabetesFaculty of MedicineKindai UniversityOsaka‐sayama, OsakaJapan
| |
Collapse
|
43
|
George MN, Leavens KF, Gadue P. Genome Editing Human Pluripotent Stem Cells to Model β-Cell Disease and Unmask Novel Genetic Modifiers. Front Endocrinol (Lausanne) 2021; 12:682625. [PMID: 34149620 PMCID: PMC8206553 DOI: 10.3389/fendo.2021.682625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/13/2021] [Indexed: 01/21/2023] Open
Abstract
A mechanistic understanding of the genetic basis of complex diseases such as diabetes mellitus remain elusive due in large part to the activity of genetic disease modifiers that impact the penetrance and/or presentation of disease phenotypes. In the face of such complexity, rare forms of diabetes that result from single-gene mutations (monogenic diabetes) can be used to model the contribution of individual genetic factors to pancreatic β-cell dysfunction and the breakdown of glucose homeostasis. Here we review the contribution of protein coding and non-protein coding genetic disease modifiers to the pathogenesis of diabetes subtypes, as well as how recent technological advances in the generation, differentiation, and genome editing of human pluripotent stem cells (hPSC) enable the development of cell-based disease models. Finally, we describe a disease modifier discovery platform that utilizes these technologies to identify novel genetic modifiers using induced pluripotent stem cells (iPSC) derived from patients with monogenic diabetes caused by heterozygous mutations.
Collapse
Affiliation(s)
- Matthew N. George
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Karla F. Leavens
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Paul Gadue
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
44
|
Sjöholm Å. GAD-65 antibodies in a case of HNF1A-Maturity-Onset Diabetes of the Young: Double diabetes? Clin Case Rep 2021; 9:e04151. [PMID: 34194751 PMCID: PMC8222641 DOI: 10.1002/ccr3.4151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/18/2021] [Accepted: 03/30/2021] [Indexed: 11/09/2022] Open
Abstract
Diabetes classification is not as defined as it used to be. A patient with one type of diabetes can have diagnostic criteria of another type, which may affect the course of the disease. Clinicians need to consider that when dealing with patients who do not fit the exact description of their diagnosed type of diabetes.
Collapse
Affiliation(s)
- Åke Sjöholm
- Division of Endocrinology and DiabetologyDepartment of Internal MedicineGävle HospitalGävleSweden
- University of GävleGävleSweden
| |
Collapse
|
45
|
London S, De Franco E, Elias-Assad G, Barhoum MN, Felszer C, Paniakov M, Weiner SA, Tenenbaum-Rakover Y. Case Report: Neonatal Diabetes Mellitus Caused by a Novel GLIS3 Mutation in Twins. Front Endocrinol (Lausanne) 2021; 12:673755. [PMID: 34093443 PMCID: PMC8169976 DOI: 10.3389/fendo.2021.673755] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/29/2021] [Indexed: 11/21/2022] Open
Abstract
Background Mutations in GLIS3 cause a rare syndrome characterized by neonatal diabetes mellitus (NDM), congenital hypothyroidism, congenital glaucoma and cystic kidneys. To date, 14 mutations in GLIS3 have been reported, inherited in an autosomal recessive manner. GLIS3 is a key transcription factor involved in β-cell development, insulin expression, and development of the thyroid, eyes, liver and kidneys. Cases We describe non-identical twins born to consanguineous parents presenting with NDM, congenital hypothyroidism, congenital glaucoma, hepatic cholestasis, cystic kidney and delayed psychomotor development. Sequence analysis of GLIS3 identified a novel homozygous nonsense mutation, c.2392C>T, p.Gln798Ter (p.Q798*), which results in an early stop codon. The diabetes was treated with a continuous subcutaneous insulin infusion pump and continuous glucose monitoring. Fluctuating blood glucose and intermittent hypoglycemia were observed on follow-up. Conclusions This report highlights the importance of early molecular diagnosis for appropriate management of NDM. We describe a novel nonsense mutation of GLIS3 causing NDM, extend the phenotype, and discuss the challenges in clinical management. Our findings provide new areas for further investigation into the roles of GLIS3 in the pathophysiology of diabetes mellitus.
Collapse
Affiliation(s)
- Shira London
- Pediatric Endocrine Institute, Ha’Emek Medical Center, Afula, Israel
| | - Elisa De Franco
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Ghadir Elias-Assad
- Pediatric Endocrine Institute, Ha’Emek Medical Center, Afula, Israel
- The Rappaport Faculty of Medicine, Technion – Institute of Technology, Haifa, Israel
| | - Marie Noufi Barhoum
- Pediatric Endocrine Institute, Ha’Emek Medical Center, Afula, Israel
- Clalit Health Services, Children Health Center, Naharia, Israel
- Faculty of Medicine, Bar-Ilan University, Zeffat, Israel
| | - Clari Felszer
- Neonatal Intensive Care Unit, Ha’Emek Medical Center, Afula, Israel
| | - Marina Paniakov
- Neonatal Intensive Care Unit, Ha’Emek Medical Center, Afula, Israel
| | - Scott A. Weiner
- Neonatal Intensive Care Unit, Ha’Emek Medical Center, Afula, Israel
| | - Yardena Tenenbaum-Rakover
- Pediatric Endocrine Institute, Ha’Emek Medical Center, Afula, Israel
- The Rappaport Faculty of Medicine, Technion – Institute of Technology, Haifa, Israel
| |
Collapse
|
46
|
Elias-Assad G, Saab R, Molnes J, Hess O, Abu-Ras R, Darawshi H, Rasmus Njølstad P, Tenenbaum-Rakover Y. Maturity onset diabetes of the young type 2 (MODY2): Insight from an extended family. Diabetes Res Clin Pract 2021; 175:108791. [PMID: 33812904 DOI: 10.1016/j.diabres.2021.108791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/18/2021] [Accepted: 03/30/2021] [Indexed: 10/21/2022]
Abstract
AIMS To assess long-term outcome of patients with maturity onset diabetes of the young, type 2 (MODY2) in a unique large cohort of patients with the same genetic and environmental background. METHODS We prospectively evaluated 162 patients aged 5 to 82 years, belonging to the same extended family living in the same village. All patients underwent molecular testing for the glucokinase (GCK) gene mutation identified in the proband, and were categorized into three groups (MODY2, type 2 diabetes and controls). RESULTS The 5.5-year-old proband had the c.1278_1286del mutation in the GCK and was diagnosed with MODY2. Forty-two out of 162 participants were positive for the mutation and 39 had type 2 diabetes. Patients were followed for a mean 10.2 ± 3.7 years (range 0-14). Mean fasting blood glucose and HbA1c increased significantly over the years in MODY2 patients (133 vs. 146 mg/dL; 6.9% vs. 8.2%, respectively). Increase in HbA1c occurred only in the obese/overweight subgroups. Twenty-five percent of MODY2 patients developed diabetes complications, all were above 40 years of age. CONCLUSIONS Although MODY2 commonly has a benign disease course, weight gain is a risk factor for diabetes complications, requiring life-long follow-up and in some patients, medical intervention.
Collapse
Affiliation(s)
- Ghadir Elias-Assad
- Pediatric Endocrine Institute, Ha'Emek Medical Center, Afula, Israel; The Rappaport Faculty of Medicine, Israel Institute of Technology, Haifa, Israel.
| | | | - Janne Molnes
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Ora Hess
- Pediatric Endocrine Institute, Ha'Emek Medical Center, Afula, Israel
| | - Rasmi Abu-Ras
- Faculty of Medicine, Bar-Ilan University, Zefat, Israel
| | | | - Pal Rasmus Njølstad
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Pediatrics and Adolescents, Haukeland University Hospital, Bergen, Norway
| | - Yardena Tenenbaum-Rakover
- Pediatric Endocrine Institute, Ha'Emek Medical Center, Afula, Israel; The Rappaport Faculty of Medicine, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
47
|
Sobczyk MK, Gaunt TR, Paternoster L. MendelVar: gene prioritization at GWAS loci using phenotypic enrichment of Mendelian disease genes. Bioinformatics 2021; 37:1-8. [PMID: 33836063 PMCID: PMC8034535 DOI: 10.1093/bioinformatics/btaa1096] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/30/2020] [Accepted: 01/08/2021] [Indexed: 11/26/2022] Open
Abstract
Motivation Gene prioritization at human GWAS loci is challenging due to linkage-disequilibrium and long-range gene regulatory mechanisms. However, identifying the causal gene is crucial to enable identification of potential drug targets and better understanding of molecular mechanisms. Mapping GWAS traits to known phenotypically relevant Mendelian disease genes near a locus is a promising approach to gene prioritization. Results We present MendelVar, a comprehensive tool that integrates knowledge from four databases on Mendelian disease genes with enrichment testing for a range of associated functional annotations such as Human Phenotype Ontology, Disease Ontology and variants from ClinVar. This open web-based platform enables users to strengthen the case for causal importance of phenotypically matched candidate genes at GWAS loci. We demonstrate the use of MendelVar in post-GWAS gene annotation for type 1 diabetes, type 2 diabetes, blood lipids and atopic dermatitis. Availability and implementation MendelVar is freely available at https://mendelvar.mrcieu.ac.uk Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- M K Sobczyk
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - T R Gaunt
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - L Paternoster
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| |
Collapse
|
48
|
Ortega MA, Fraile-Martínez O, Pekarek L, Alvarez-Mon MA, Asúnsolo Á, Sanchez-Trujillo L, Coca S, Buján J, Álvarez-Mon M, García-Honduvilla N, Sainz F. Defective expression of the peroxisome regulators PPARα receptors and lysogenesis with increased cellular senescence in the venous wall of chronic venous disorder. Histol Histopathol 2021; 36:547-558. [PMID: 33645625 DOI: 10.14670/hh-18-322] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The pathogenesis of chronic venous disorder (CVeD) remains partially understood. A marked wall remodeling has been shown with potential accelerated tissue senescence. We have investigated the expression of peroxisome proliferator-activated receptor (PPAR) isoforms transcription factor EB (TFEB) as regulatory molecules of cellular homeostasis and makers of peroxisomal and lysosomal biogenesis. We have also quantified p16 expression as a cellular senescence marker. In specimens of maior safena vein from 35 CVeD and 27 healthy venous controls (HV), we studied the expression of PPAR-α, PPAR-β/δ, PPAR-γ, TFEB and p16 by RT-qPCR and immunohistochemical techniques. We have demonstrated a reduced gene and protein expression of the PPAR-α and PPAR-β/δ isoform as well as that of TFEB in the venous wall of CVeD patients, suggesting an altered peroxisomal and lysosomal biogenesis associated with an increased cellular senescence shown by increased p16 expression.
Collapse
Affiliation(s)
- Miguel A Ortega
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain.,Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain.,Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Leonel Pekarek
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Miguel A Alvarez-Mon
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain.,Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Ángel Asúnsolo
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain.,Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcala, Alcala de Henares, Madrid, Spain
| | - Lara Sanchez-Trujillo
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain.,Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain.,Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Santiago Coca
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain.,Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Julia Buján
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain.,Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain.
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain.,Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain.,Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine, University Hospital Príncipe de Asturias, (CIBEREHD), Alcalá de Henares, Madrid, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain.,Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Felipe Sainz
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain.,Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcala, Alcala de Henares, Madrid, Spain.,Angiology and Vascular Surgery Service, Central University Hospital of Defence-UAH Madrid, Spain
| |
Collapse
|
49
|
Hou J, Zhou X, Wang P, Zhao C, Qin Y, Liu F, Yu L, Xu H. An Integrative Pharmacology-Based Approach for Evaluating the Potential Effects of Purslane Seed in Diabetes Mellitus Treatment Using UHPLC-LTQ-Orbitrap and TCMIP V2.0. Front Pharmacol 2021; 11:593693. [PMID: 33603663 PMCID: PMC7884824 DOI: 10.3389/fphar.2020.593693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/09/2020] [Indexed: 01/09/2023] Open
Abstract
Portulaca oleracea L., known as the “vegetable for long life,” is an annual succulent herb that is widely distributed worldwide. Many clinical and experimental studies have demonstrated that purslane seed (MCXZ) can be used as an adjunctive and alternative therapy for the treatment of diabetes mellitus (DM). However, the underlying active constituents and pharmacological mechanisms through which MCXZ exerts effects in DM remain unclear. In the present study, we confirmed that MCXZ treatment resulted in hypoglycemic activity, lowering the fasting blood glucose and glycated hemoglobin levels in streptozotocin-induced diabetic mice. Then, ultra-high-pressure liquid chromatography coupled with linear ion trap-Orbitrap tandem mass spectrometry was used to systematically analyze the chemical profile of MCXZ, resulting in the identification of 84 constituents, including 31 organic acids and nine flavonoids. Finally, the Integrative Pharmacology-based Research Platform of Traditional Chinese Medicine was employed to analyze the key active components of MCXZ and the molecular mechanisms through which these components acted in DM. Ten key active compounds were identified based on the topological importance of their corresponding putative targets within the known DM-associated therapeutic target network of known MCXZ putative targets. Functionally, these candidate targets play critical anti-hyperlipidemia, anti-hyperglycemia, immunity regulation, and inflammatory roles involving DM-related pathways, such as the vascular endothelial growth factor (VEGF) signaling pathway and Fc gamma R-mediated phagocytosis, which indicated that MCXZ exhibited anti-diabetic activity through multi-faced actions.
Collapse
Affiliation(s)
- Jinli Hou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiang Zhou
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Ping Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chunhui Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuewen Qin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Feng Liu
- Medical College, Shaanxi Institute of International Trade and Commerce, Xianyang, China
| | - Liping Yu
- Guangzhou Zhongda Pharmaceutical Development Co. Ltd., Guangzhou, China
| | - Haiyu Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.,Medical College, Shaanxi Institute of International Trade and Commerce, Xianyang, China
| |
Collapse
|
50
|
Bourgeois S, Sawatani T, Van Mulders A, De Leu N, Heremans Y, Heimberg H, Cnop M, Staels W. Towards a Functional Cure for Diabetes Using Stem Cell-Derived Beta Cells: Are We There Yet? Cells 2021; 10:cells10010191. [PMID: 33477961 PMCID: PMC7835995 DOI: 10.3390/cells10010191] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus is a pandemic metabolic disorder that results from either the autoimmune destruction or the dysfunction of insulin-producing pancreatic beta cells. A promising cure is beta cell replacement through the transplantation of islets of Langerhans. However, donor shortage hinders the widespread implementation of this therapy. Human pluripotent stem cells, including embryonic stem cells and induced pluripotent stem cells, represent an attractive alternative beta cell source for transplantation. Although major advances over the past two decades have led to the generation of stem cell-derived beta-like cells that share many features with genuine beta cells, producing fully mature beta cells remains challenging. Here, we review the current status of beta cell differentiation protocols and highlight specific challenges that are associated with producing mature beta cells. We address the challenges and opportunities that are offered by monogenic forms of diabetes. Finally, we discuss the remaining hurdles for clinical application of stem cell-derived beta cells and the status of ongoing clinical trials.
Collapse
Affiliation(s)
- Stephanie Bourgeois
- Beta Cell Neogenesis (BENE) Research Group, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (S.B.); (A.V.M.); (N.D.L.); (Y.H.); (H.H.)
| | - Toshiaki Sawatani
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, 1070 Brussels, Belgium; (T.S.); (M.C.)
| | - Annelore Van Mulders
- Beta Cell Neogenesis (BENE) Research Group, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (S.B.); (A.V.M.); (N.D.L.); (Y.H.); (H.H.)
| | - Nico De Leu
- Beta Cell Neogenesis (BENE) Research Group, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (S.B.); (A.V.M.); (N.D.L.); (Y.H.); (H.H.)
- Department of Endocrinology, University Hospital Brussels, 1090 Brussels, Belgium
- Department of Endocrinology, ASZ Aalst, 9300 Aalst, Belgium
| | - Yves Heremans
- Beta Cell Neogenesis (BENE) Research Group, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (S.B.); (A.V.M.); (N.D.L.); (Y.H.); (H.H.)
| | - Harry Heimberg
- Beta Cell Neogenesis (BENE) Research Group, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (S.B.); (A.V.M.); (N.D.L.); (Y.H.); (H.H.)
| | - Miriam Cnop
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, 1070 Brussels, Belgium; (T.S.); (M.C.)
- Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Willem Staels
- Beta Cell Neogenesis (BENE) Research Group, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (S.B.); (A.V.M.); (N.D.L.); (Y.H.); (H.H.)
- Service of Pediatric Endocrinology, Department of Pediatrics, KidZ Health Castle, Universitair Ziekenhuis Brussel (UZ Brussel), 1090 Brussels, Belgium
- Correspondence: ; Tel.: +32-0-24774473
| |
Collapse
|