1
|
Bhattacharya S, Fernandez CJ, Kamrul-Hasan ABM, Pappachan JM. Monogenic diabetes: An evidence-based clinical approach. World J Diabetes 2025; 16:104787. [DOI: 10.4239/wjd.v16.i5.104787] [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: 12/31/2024] [Revised: 02/20/2025] [Accepted: 03/11/2025] [Indexed: 04/25/2025] Open
Abstract
Monogenic diabetes is a heterogeneous disorder characterized by hyperglycemia arising from defects in a single gene. Maturity-onset diabetes of the young (MODY) is the most common type with 14 subtypes, each linked to specific mutations affecting insulin synthesis, secretion and glucose regulation. Common traits across MODY subtypes include early-onset diabetes, a family history of autosomal dominant diabetes, lack of features of insulin resistance, and absent islet cell autoimmunity. Many cases are misdiagnosed as type 1 and type 2 diabetes mellitus. Biomarkers and scoring systems can help identify candidates for genetic testing. GCK-MODY, a common subtype, manifests as mild hyperglycemia and doesn’t require treatment except during pregnancy. In contrast, mutations in HNF4A, HNF1A, and HNF1B genes lead to progressive beta-cell failure and similar risks of complications as type 2 diabetes mellitus. Neonatal diabetes mellitus (NDM) is a rare form of monogenic diabetes that usually presents within the first six months. Half of the cases are lifelong, while others experience transient remission. Permanent NDM is most commonly due to activating mutations in genes encoding the adenosine triphosphate-sensitive potassium channel (KCNJ11 or ABCC8) and can be transitioned to sulfonylurea after confirmation of diagnosis. Thus, in many cases, monogenic diabetes offers an opportunity to provide precision treatment. The scope has broadened with next-generation sequencing (NGS) technologies, replacing older methods like Sanger sequencing. NGS can be for targeted gene panels, whole-exome sequencing (WES), or whole-genome sequencing. Targeted gene panels offer specific information efficiently, while WES provides comprehensive data but comes with bioinformatic challenges. The surge in testing has also led to an increase in variants of unknown significance (VUS). Deciding whether VUS is disease-causing or benign can be challenging. Computational models, functional studies, and clinical knowledge help to determine pathogenicity. Advances in genetic testing technologies offer hope for improved diagnosis and personalized treatment but also raise concerns about interpretation and ethics.
Collapse
Affiliation(s)
| | - Cornelius J Fernandez
- Department of Endocrinology and Metabolism, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston PE21 9QS, Lincolnshire, United Kingdom
| | | | - Joseph M Pappachan
- Faculty of Science, Manchester Metropolitan University, Manchester M15 6BH, Greater Manchester, United Kingdom
- Department of Endocrinology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
2
|
Russ-Silsby J, Lee Y, Rajesh V, Amoli M, Mirhosseini NA, Godbole T, Johnson MB, Ibarra DE, Sun H, Krentz NAJ, Wakeling MN, Flanagan SE, Hattersley AT, Gloyn AL, De Franco E. Complete Loss of PAX4 causes Transient Neonatal Diabetes in Humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.01.25324926. [PMID: 40236391 PMCID: PMC11998800 DOI: 10.1101/2025.04.01.25324926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Gene discovery studies in individuals with diabetes diagnosed within 6 months of life (neonatal diabetes, NDM) can provide unique insights into the development and function of human pancreatic beta-cells. We describe the identification of homozygous PAX4 loss-of-function variants in 2 unrelated individuals with NDM: a p.(Arg126*) stop-gain variant and a c.-352_104del deletion affecting the first 4 PAX4 exons. We confirmed the p.(Arg126*) variant causes nonsense mediated decay in CRISPR-edited human induced pluripotent stem cell (iPSC)-derived pancreatic endoderm cells. Integrated analysis of CUT&RUN and RNA-sequencing in PAX4-depleted islet cell models identified genes directly regulated by PAX4 involved in both pancreatic islet development and glucose-stimulated insulin secretion. Both probands had transient NDM which remitted in early infancy but relapsed between the ages of 2 and 7 years, demonstrating that in contrast to mouse models, PAX4 is not essential for the development of human pancreatic beta-cells.
Collapse
Affiliation(s)
- James Russ-Silsby
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Yunkyeong Lee
- Division of Endocrinology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Varsha Rajesh
- Division of Endocrinology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Mahsa Amoli
- Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Iran
| | | | | | - Matthew B. Johnson
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Dora E. Ibarra
- Division of Endocrinology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Han Sun
- Division of Endocrinology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Nicole A. J. Krentz
- Division of Endocrinology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Matthew N. Wakeling
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Sarah E. Flanagan
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Andrew T. Hattersley
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Anna L. Gloyn
- Division of Endocrinology, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
- Stanford Diabetes Research Center, School of Medicine, Stanford University, Stanford, CA, USA
| | - Elisa De Franco
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| |
Collapse
|
3
|
Görükmez O, Görükmez Ö, Topak A, Arsoy HA. Clinical Exome Sequencing in Pediatric Patients. Cureus 2025; 17:e80330. [PMID: 40206918 PMCID: PMC11980008 DOI: 10.7759/cureus.80330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
INTRODUCTION The development of genomic sequencing techniques has led to the effective diagnosis of genetic diseases. In this study, clinical exome sequencing (CES) results applied to genetic disorders are reported. METHODS The CES results of pediatric patients with different system involvements and whose complaints were thought to be of genetic origin were evaluated retrospectively. RESULTS Significant variants associated with complaints were detected in 41 (60%) of 68 patients. Copy number variations were detected in two patients, and single nucleotide variants (SNVs) were detected in the other 39 patients. A total of 46 SNVs were detected in these 39 patients. Sixteen of the detected SNVs were previously reported in the literature, but 30 were novel. CONCLUSIONS This study shows that CES can provide a high diagnosis rate (60%) in childhood genetic diseases. Novel mutations (30) have contributed to the mutation profiles of genetic disorders.
Collapse
Affiliation(s)
- Orhan Görükmez
- Medical Genetics, Bursa Yüksek İhtisas Training and Research Hospital, Bursa, TUR
| | - Özlem Görükmez
- Medical Genetics, Bursa Yüksek İhtisas Training and Research Hospital, Bursa, TUR
| | - Ali Topak
- Medical Genetics, Bursa State Hospital, Bursa, TUR
| | | |
Collapse
|
4
|
Luckett AM, Hawkes G, Green HD, De Franco E, Hagopian WA, Roep BO, Weedon MN, Oram RA, Johnson MB. Type 1 Diabetes Genetic Risk Contributes to Phenotypic Presentation in Monogenic Autoimmune Diabetes. Diabetes 2025; 74:243-248. [PMID: 39531505 PMCID: PMC11755682 DOI: 10.2337/db24-0485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
ARTICLE HIGHLIGHTS There is variability in early-onset autoimmune diabetes presentation in individuals with monogenic autoimmunity; the mechanism(s) underlying this is unclear. We examined whether type 1 diabetes (T1D) polygenic risk contributes to clinical phenotype in monogenic autoimmune diabetes. Individuals with monogenic autoimmune diabetes had higher T1D genetic risk scores compared with control cohorts, driven largely by increased presence of T1D-risk DR3-DQ2 haplotype. Established T1D polygenic risk alleles, particularly class II HLA genes, contribute to clinical presentation in monogenic autoimmunity.
Collapse
Affiliation(s)
- Amber M. Luckett
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
| | - Gareth Hawkes
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
| | - Harry D. Green
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
| | - Elisa De Franco
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
| | - William A. Hagopian
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Bart O. Roep
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Michael N. Weedon
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
| | - Richard A. Oram
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
- Royal Devon University Healthcare NHS Foundation Trust, Exeter, U.K
| | - Matthew B. Johnson
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
| |
Collapse
|
5
|
Minniakhmetov IR, Khusainova RI, Laptev DN, Yalaev BI, Karpova YS, Deev RV, Salakhov RR, Panteleev DD, Smirnov KV, Melnichenko GA, Shestakova MV, Mokrysheva NG. Genetic Structure of Hereditary Forms of Diabetes Mellitus in Russia. Int J Mol Sci 2025; 26:740. [PMID: 39859454 PMCID: PMC11766241 DOI: 10.3390/ijms26020740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Analyzing the genetic architecture of hereditary forms of diabetes in different populations is a critical step toward optimizing diagnostic and preventive algorithms. This requires consideration of regional and population-specific characteristics, including the spectrum and frequency of pathogenic variants in targeted genes. As part of this study, we used a custom-designed NGS panel to screen for mutations in 28 genes associated with the pathogenesis of hereditary diabetes mellitus in 506 unrelated patients from Russia. The study identified 180 pathogenic or likely pathogenic variants across 13 genes (GCK, HNF1A, HNF1B, HNF4A, ABCC8, INS, INSR, KCNJ11, PAX4, PDX1, ZFP57, BLK, WFS1), representing 46.44% of the analyzed cohort (235 individuals). The glucokinase gene (GCK) had the highest number of identified variants, with 111 variants detected in 161 patients, 20 of which were identified for the first time. In the tissue-specific transcription factor genes HNF1A, HNF4A, and HNF1B, 34 variants were found in 38 patients, including 13 that were previously unreported. Seventeen variants were identified in the ABCC8 gene, which encodes the ATP-binding cassette transporter 8 of subfamily C, each found in a different patient; four of these were novel discoveries. Nine pathogenic or likely pathogenic variants were identified in the insulin gene (INS) and its receptor gene (INSR), including four previously unreported variants. Additionally, we identified 10 previously unreported variants in six other genes among 11 patients. Variants in the genes GCK, HNF1A, HNF1B, HNF4A, ABCC8, INS, and INSR were the main contributors to the genetic pathogenesis of hereditary diabetes mellitus in the Russian cohort. These findings enhance our understanding of the molecular mechanisms underlying the disease and provide a solid basis for future studies aimed at improving diagnostic accuracy and advancing personalized therapeutic strategies. This knowledge provides a foundation for developing region-specific genetic testing algorithms and personalized therapeutic strategies, which are critical for future initiatives in precision medicine.
Collapse
Affiliation(s)
- Ildar R. Minniakhmetov
- Endocrinology Research Center, Moscow 117292, Russia; (R.I.K.); (D.N.L.); (B.I.Y.); (Y.S.K.); (R.V.D.); (R.R.S.); (D.D.P.); (K.V.S.); (G.A.M.); (M.V.S.); (N.G.M.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Tsoy UA, Sokolnikova PS, Kravchuk EN, Ryazanov PA, Kozyreva AA, Fomicheva YV, Aramisova LS, Karonova TL, Kostareva AA, Grineva E. A Comprehensive Target Panel Allows to Extend the Genetic Spectrum of Neuroendocrine Tumors. Neuroendocrinology 2024; 115:381-401. [PMID: 39536727 PMCID: PMC12169807 DOI: 10.1159/000542223] [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: 04/09/2024] [Accepted: 09/17/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Neuroendocrine tumors (NETs) frequently have a genetic basis, and the range of genes implicated in NET development continues to expand. Application of targeted gene panels (TGPs) in next-generation sequencing is a central strategy for elucidating novel variants associated with NET development. METHODS In this study, we conducted comprehensive molecular genetic analyses using TGP on a cohort of 93 patients diagnosed with various NETs subtypes, mainly accompanied by various endocrine syndromes: insulinoma (n = 26), pheochromocytoma and paraganglioma (PPGL) (n = 38), parathyroid adenoma (n = 18, including three with insulinoma), and NETs of other locations (n = 14). The TGP encompassed genes linked to diverse NETs and other hereditary endocrine disorders, with subsequent variant classification according to the American College of Medical Genetics and Genomics guidelines. RESULTS Among the identified variants, 20 were found in genes previously linked to specific tumor types, and 10 were found in genes with a limited likelihood and unclear molecular mecanisms of association with observed NETs. Remarkably, 13 variants were discovered in genes not previously associated with the NETs observed in our patients. These genes, such as ABCC8, KCNJ11, KLF11, HABP2, and APC, were implicated in insulinoma; ZNRF3, GNAS, and KCNJ5 were linked with PPGL; parathyroid adenomas were related to variants in SDHB and TP53; while NETs of other locations displayed variants in APC and ABCC8. CONCLUSION Our study demonstrates that utilizing broad TGP in examining patients with various functioning NETs facilitates the identification of new germinal variants in genes that may contribute to the diseases. The verification of revealed findings requires research in vaster sample.
Collapse
Affiliation(s)
- Uliana A. Tsoy
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Polina S. Sokolnikova
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Ekaterina N. Kravchuk
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Pavel A. Ryazanov
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Alexandra A. Kozyreva
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Yulia V. Fomicheva
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Liana S. Aramisova
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Tatiana L. Karonova
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Anna A. Kostareva
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
- Department of Women’s and Children’s Health, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Elena Grineva
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| |
Collapse
|
7
|
Tanaka S, Akagawa H, Azuma K, Higuchi S, Ujiie A, Hashimoto K, Iwasaki N. High prevalence of copy number variations in the Japanese participants with suspected MODY. Clin Genet 2024; 106:293-304. [PMID: 38733153 DOI: 10.1111/cge.14544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
Maturity-Onset Diabetes of the Young (MODY) is a diabetes mellitus subtype caused by a single gene. The detection rate of the responsible gene is 27% in the United Kingdom, indicating that the causative gene remains unknown in the majority of clinically diagnosed MODY cases. To improve the detection rate, we applied comprehensive genetic testing using whole exome sequencing (WES) followed by Multiplex Ligation-dependent Probe Amplification (MLPA) and functional analyses. Twenty-one unrelated Japanese participants with MODY were enrolled in the study. To detect copy number variations (CNVs), WES was performed first, followed by MLPA analysis for participants who were negative on the basis of WES. Undetermined variants were analyzed according to their functional properties. WES identified 7 pathogenic and 3 novel likely pathogenic variants in the 21 participants. Functional analyses revealed that 1 in 3 variants was pathogenic. MLPA analysis applied to the remaining 13 undetermined samples identified 4 cases with pathogenic CNVs: 3 in HNF4A and 1 in HNF1B. Pathogenic variants were identified in 12 participants (12/21, 57.1%) - relatively high rate reported to date. Notably, one-third of the participants had CNVs in HNF4A or HNF1B, indicating a limitation of WES-only screening.
Collapse
Affiliation(s)
- Satoshi Tanaka
- Institute for Comprehensive Medical Sciences, Tokyo Women's Medical University, Tokyo, Japan
- Diabetes and Metabolism, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Hiroyuki Akagawa
- Institute for Comprehensive Medical Sciences, Tokyo Women's Medical University, Tokyo, Japan
- Department of Neurosurgery, Tokyo Women's Medical University Adachi Medical Center, Tokyo, Japan
| | - Kenkou Azuma
- Institute for Comprehensive Medical Sciences, Tokyo Women's Medical University, Tokyo, Japan
| | - Sayaka Higuchi
- Institute for Comprehensive Medical Sciences, Tokyo Women's Medical University, Tokyo, Japan
| | - Atsushi Ujiie
- Department of Diabetes, Endocrinology and Hematology, Dokkyo Medical University Saitama Medical Center, Saitama, Japan
| | - Koshi Hashimoto
- Department of Diabetes, Endocrinology and Hematology, Dokkyo Medical University Saitama Medical Center, Saitama, Japan
| | - Naoko Iwasaki
- Institute for Comprehensive Medical Sciences, Tokyo Women's Medical University, Tokyo, Japan
- Diabetes and Metabolism, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
- Division of Diabetes, Endocrinology and Metabolism, Tokyo Women's Medical University Yachiyo Medical Center, Chiba, Japan
| |
Collapse
|
8
|
Wright CF, Sharp LN, Jackson L, Murray A, Ware JS, MacArthur DG, Rehm HL, Patel KA, Weedon MN. Guidance for estimating penetrance of monogenic disease-causing variants in population cohorts. Nat Genet 2024; 56:1772-1779. [PMID: 39075210 DOI: 10.1038/s41588-024-01842-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/24/2024] [Indexed: 07/31/2024]
Abstract
Penetrance is the probability that an individual with a pathogenic genetic variant develops a specific disease. Knowing the penetrance of variants for monogenic disorders is important for counseling of individuals. Until recently, estimates of penetrance have largely relied on affected individuals and their at-risk family members being clinically referred for genetic testing, a 'phenotype-first' approach. This approach substantially overestimates the penetrance of variants because of ascertainment bias. The recent availability of whole-genome sequencing data in individuals from very-large-scale population-based cohorts now allows 'genotype-first' estimates of penetrance for many conditions. Although this type of population-based study can underestimate penetrance owing to recruitment biases, it provides more accurate estimates of penetrance for secondary or incidental findings. Here, we provide guidance for the conduct of penetrance studies to ensure that robust genotypes and phenotypes are used to accurately estimate penetrance of variants and groups of similarly annotated variants from population-based studies.
Collapse
Affiliation(s)
- Caroline F Wright
- Department of Clinical and Biomedical Sciences, Medical School, University of Exeter, Exeter, UK.
| | - Luke N Sharp
- Department of Clinical and Biomedical Sciences, Medical School, University of Exeter, Exeter, UK
| | - Leigh Jackson
- Department of Clinical and Biomedical Sciences, Medical School, University of Exeter, Exeter, UK
| | - Anna Murray
- Department of Clinical and Biomedical Sciences, Medical School, University of Exeter, Exeter, UK
| | - James S Ware
- National Heart and Lung Institute and MRC Laboratory of Medical Sciences, Imperial College London, London, UK
- Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
- Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel G MacArthur
- Centre for Population Genomics, Garvan Institute of Medical Research and UNSW Sydney, Sydney, New South Wales, Australia
- Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Heidi L Rehm
- Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kashyap A Patel
- Department of Clinical and Biomedical Sciences, Medical School, University of Exeter, Exeter, UK
| | - Michael N Weedon
- Department of Clinical and Biomedical Sciences, Medical School, University of Exeter, Exeter, UK.
| |
Collapse
|
9
|
Alwithenani R, Alzahrani J, Allugmani E, Hakami F. Co-inheritance of Rare Variants of Maturity-Onset Diabetes of the Young (MODY): A Case Report and Review of the Literature. Cureus 2024; 16:e69039. [PMID: 39391454 PMCID: PMC11464862 DOI: 10.7759/cureus.69039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Maturity onset diabetes of the young (MODY) is a rare, monogenic autosomal dominant form of diabetes that is characterized by early-onset, non-insulin-dependent hyperglycemia, strong family history, and is often misdiagnosed as type 1 or type 2 diabetes. Co-inheritance of multiple MODY genes, however, is rare. We describe here a case of MODY involving co-inherited rare variants in the ABCC8 and B-lymphocyte kinase (BLK) genes. A 55-year-old non-obese man with a past medical history of dyslipidemia and premature ischemic heart disease was initially misdiagnosed with type 2 diabetes for more than 18 years. He is a smoker with a strong family history of diabetes affecting both of his parents and most of his siblings. Despite treatment with different oral antihyperglycemics, his diabetes remained uncontrolled with glycated hemoglobin (HBA1c) between 8 and 10% until the addition of gliclazide, which improved his HBA1c to 5.7%. Based on all the previous information, MODY was suspected, and genetic testing was done, which showed rare variants in the BLK and ABCC8 genes and suggested a co-occurrence of MODY11 and MODY12. This case highlights the importance of accurate genetic testing, which is crucial for proper MODY subtyping, enabling tailored treatment strategies and potentially improving patient outcomes. Moreover, the consistent presence of the BLK gene variant in limited cases of co-inheritance raises questions about its causative role in MODY, suggesting a need for further investigation into its clinical significance.
Collapse
Affiliation(s)
- Raad Alwithenani
- Department of Medicine, Division of Endocrinology and Diabetes, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Jeddah, SAU
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Jeddah, SAU
| | - Jehad Alzahrani
- Department of Medicine, Division of Endocrinology and Diabetes, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Jeddah, SAU
| | - Ebtesam Allugmani
- Department of Medicine, Division of Endocrinology and Diabetes, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Jeddah, SAU
| | - Fahad Hakami
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Jeddah, SAU
| |
Collapse
|
10
|
Peghinelli VV, De Sibio MT, Depra IDC, Teles Bezerra MG, Sakalem ME, Júnior AFDM, da Rocha PB, Tilli HP, Gonçalves BM, Vieira EM, Lourenço MM, Nogueira CR. MODY calculator applied in patients with clinical diagnosis of type 1 diabetes mellitus: Is a higher cutoff needed? Heliyon 2024; 10:e36006. [PMID: 39224250 PMCID: PMC11367115 DOI: 10.1016/j.heliyon.2024.e36006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/14/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Aim This study aimed to evaluate the mean post-test probability (PTP) of the Maturity-onset diabetes of the young (MODY) calculator in a multiethnic cohort of patients previously diagnosed with type 1 diabetes (T1DM). Materials and methods The MODY probability calculator proposed by Shields and colleagues (2012) was applied to 117 patients from a T1DM outpatient clinic at a tertiary hospital in Brazil. Additionally, two exons of the HNF1A gene were sequenced in eight patients who hadn't received insulin treatment within six months after the diagnosis. Results 17.1 % of patients achieved PTP >10 %; 11.1 % achieved PTP >25 % (and all patients >30 %), and 7.7 % achieved PTP >40 %. Among the patients who were selected for genetic sequencing, 100 % presented PTP >30 %, with 66.6 % achieving PTP >40 % and 41.6 % achieving PTP >75 %. These cutoffs are as suggested for the Brazilian population, according to previous investigations. No mutation was observed in the sequenced exons. Conclusion Considering that only around 10 % of the evaluated cases achieved PTP >30 %, it is highly probable that the most suitable cutoff to select patients for genetic sequencing in a Brazilian cohort of T1DM is higher than the cutoff used in Caucasian populations.
Collapse
Affiliation(s)
| | - Maria Teresa De Sibio
- Departament of Internal Medicine, São Paulo State University (UNESP), Medical School, Botucatu, Sao Paulo, Brazil
| | - Igor de Carvalho Depra
- Departament of Internal Medicine, São Paulo State University (UNESP), Medical School, Botucatu, Sao Paulo, Brazil
| | - Milena Gurgel Teles Bezerra
- Grupo de Diabetes Monogênico (Monogenic Diabetes Group), Unidade de Endocrinologia, Genética (LIM25), Unidade de Diabetes, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, 01246-903, Brazil
| | - Marna Eliana Sakalem
- Departament of Anatomy, State University of Londrina – UEL, Londrina, PR, Brazil
| | | | - Paula Barreto da Rocha
- Departament of Internal Medicine, São Paulo State University (UNESP), Medical School, Botucatu, Sao Paulo, Brazil
| | - Helena Paim Tilli
- Departament of Internal Medicine, São Paulo State University (UNESP), Medical School, Botucatu, Sao Paulo, Brazil
| | - Bianca Mariani Gonçalves
- Departament of Internal Medicine, São Paulo State University (UNESP), Medical School, Botucatu, Sao Paulo, Brazil
| | - Ester Mariane Vieira
- Departament of Internal Medicine, São Paulo State University (UNESP), Medical School, Botucatu, Sao Paulo, Brazil
| | - Mariana Menezes Lourenço
- Departament of Internal Medicine, São Paulo State University (UNESP), Medical School, Botucatu, Sao Paulo, Brazil
| | - Célia Regina Nogueira
- Departament of Internal Medicine, São Paulo State University (UNESP), Medical School, Botucatu, Sao Paulo, Brazil
| |
Collapse
|
11
|
Asgarian S, Lanjanian H, Rahimipour Anaraki S, Hadaegh F, Moazzam-Jazi M, Najd-Hassan-Bonab L, Masjoudi S, Zahedi AS, Zarkesh M, Shalbafan B, Akbarzadeh M, Tehrani Fateh S, Khalili D, Momenan A, Sarbazi N, Hedayati M, Azizi F, Daneshpour MS. Examining the clinical and genetic spectrum of maturity-onset diabetes of the young (MODY) in Iran. Sci Rep 2024; 14:19860. [PMID: 39191897 PMCID: PMC11349921 DOI: 10.1038/s41598-024-70864-y] [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: 02/04/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024] Open
Abstract
Maturity-onset diabetes of the young (MODY) is an uncommon monogenic type of diabetes mellitus. Detecting genetic variants for MODY is a necessity for precise diagnosis and treatment. The majority of MODY genetic predisposition has been documented in European populations and a lack of information is present in Iranians which leads to misdiagnosis as a consequence of defects in unknown variants. In this study, using genetic variant information of 20,002 participants from the family-based TCGS (Tehran Cardiometabolic Genetic Study) cohort, we evaluated the genetic spectrum of MODY in Iran. We concentrated on previously discovered MODY-causing genes. Genetic variants were evaluated for their pathogenicity. We discovered 6 variants that were previously reported in the ClinVar as pathogenic/likely pathogenic (P/LP) for MODY in 45 participants from 24 families (INS in 21 cases, GCK in 13, HNF1B in 8, HNF4A, HNF1A, and CEL in 1 case). One potential MODY variant with Uncertain Risk Allele in ClinVar classification was also identified, which showed complete disease penetrance (100%) in four subjects from one family. This is the first family-based study to define the genetic spectrum and estimate the prevalence of MODY in Iran. The discovered variants need to be investigated by additional studies.
Collapse
Affiliation(s)
- Sara Asgarian
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 19195-4763, Tehran, Iran
| | - Hossein Lanjanian
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 19195-4763, Tehran, Iran.
| | | | - Farzad Hadaegh
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Moazzam-Jazi
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 19195-4763, Tehran, Iran
| | - Leila Najd-Hassan-Bonab
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 19195-4763, Tehran, Iran
| | - Sajedeh Masjoudi
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 19195-4763, Tehran, Iran
| | - Asiyeh Sadat Zahedi
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 19195-4763, Tehran, Iran
| | - Maryam Zarkesh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 19195-4763, Tehran, Iran
| | - Bita Shalbafan
- Clinical Research Development Center of Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Akbarzadeh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 19195-4763, Tehran, Iran
| | | | - Davood Khalili
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirabbas Momenan
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Narges Sarbazi
- Prevention of Metabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 19195-4763, Tehran, Iran
| | - Fereidoun Azizi
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 19195-4763, Tehran, Iran
| | - Maryam S Daneshpour
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, P.O. Box 19195-4763, Tehran, Iran.
| |
Collapse
|
12
|
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
|
13
|
Peixoto-Barbosa R, Calliari LE, Crispim F, Moisés RS, Dib SA, Reis AF, Giuffrida FMA. Clinical screening for GCK-MODY in 2,989 patients from the Brazilian Monogenic Diabetes Study Group (BRASMOD) and the Brazilian Type 1 Diabetes Study Group (BrazDiab1SG). ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e230314. [PMID: 39420902 PMCID: PMC11326741 DOI: 10.20945/2359-4292-2023-0314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 03/03/2024] [Indexed: 10/19/2024]
Abstract
Objectives To evaluate the accuracy of routinely available parameters in screening for GCK maturity-onset diabetes of the young (MODY), leveraging data from two large cohorts - one of patients with GCK-MODY and the other of patients with type 1 diabetes (T1D). Materials and methods The study included 2,687 patients with T1D, 202 patients with clinical features of MODY but without associated genetic variants (NoVar), and 100 patients with GCK-MODY (GCK). Area under the receiver-operating characteristic curve (ROC-AUC) analyses were used to assess the performance of each parameter - both alone and incorporated into regression models - in discriminating between groups. Results The best parameter discriminating between GCK-MODY and T1D was a multivariable model comprising glycated hemoglobin (HbA1c), fasting plasma glucose, age at diagnosis, hypertension, microvascular complications, previous diabetic ketoacidosis, and family history of diabetes. This model had a ROC-AUC value of 0.980 (95% confidence interval [CI] 0.974-0.985) and positive (PPV) and negative (NPV) predictive values of 43.74% and 100%, respectively. The best model discriminating between GCK and NoVar included HbA1c, age at diagnosis, hypertension, and triglycerides and had a ROC-AUC value of 0.850 (95% CI 0.783-0.916), PPV of 88.36%, and NPV of 97.7%; however, this model was not significantly different from the others. A novel GCK variant was also described in one individual with MODY (7-44192948-T-C, p.Ser54Gly), which showed evidence of pathogenicity on in silico prediction tools. Conclusions This study identified a highly accurate (98%) composite model for differentiating GCK-MODY and T1D. This model may help clinicians select patients for genetic evaluation of monogenic diabetes, enabling them to implement correct treatment without overusing limited resources.
Collapse
Affiliation(s)
- Renata Peixoto-Barbosa
- Universidade Federal de São PauloSão PauloSPBrasil Disciplina de Endocrinologia, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
- Departamento de Ciências da VidaUniversidade do Estado da BahiaSalvadorBABrasil Departamento de Ciências da Vida, Universidade do Estado da Bahia (Uneb), Salvador, BA, Brasil
| | - Luis Eduardo Calliari
- Departamento de PediatriaFaculdade de Ciências MédicasSanta Casa de Misericórdia de São PauloSão PauloSPBrasil Departamento de Pediatria, Faculdade de Ciências Médicas da Santa Casa de Misericórdia de São Paulo, São Paulo, SP, Brasil
| | - Felipe Crispim
- Universidade Federal de São PauloSão PauloSPBrasil Disciplina de Endocrinologia, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Regina S. Moisés
- Universidade Federal de São PauloSão PauloSPBrasil Disciplina de Endocrinologia, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Sergio A. Dib
- Universidade Federal de São PauloSão PauloSPBrasil Disciplina de Endocrinologia, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - André F. Reis
- Universidade Federal de São PauloSão PauloSPBrasil Disciplina de Endocrinologia, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Fernando M. A. Giuffrida
- Universidade Federal de São PauloSão PauloSPBrasil Disciplina de Endocrinologia, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
- Departamento de Ciências da VidaUniversidade do Estado da BahiaSalvadorBABrasil Departamento de Ciências da Vida, Universidade do Estado da Bahia (Uneb), Salvador, BA, Brasil
| |
Collapse
|
14
|
Kuznetsova KG, Vašíček J, Skiadopoulou D, Molnes J, Udler M, Johansson S, Njølstad PR, Manning A, Vaudel M. Bioinformatics pipeline for the systematic mining genomic and proteomic variation linked to rare diseases: The example of monogenic diabetes. PLoS One 2024; 19:e0300350. [PMID: 38635808 PMCID: PMC11025945 DOI: 10.1371/journal.pone.0300350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 02/23/2024] [Indexed: 04/20/2024] Open
Abstract
Monogenic diabetes is characterized as a group of diseases caused by rare variants in single genes. Like for other rare diseases, multiple genes have been linked to monogenic diabetes with different measures of pathogenicity, but the information on the genes and variants is not unified among different resources, making it challenging to process them informatically. We have developed an automated pipeline for collecting and harmonizing data on genetic variants linked to monogenic diabetes. Furthermore, we have translated variant genetic sequences into protein sequences accounting for all protein isoforms and their variants. This allows researchers to consolidate information on variant genes and proteins linked to monogenic diabetes and facilitates their study using proteomics or structural biology. Our open and flexible implementation using Jupyter notebooks enables tailoring and modifying the pipeline and its application to other rare diseases.
Collapse
Affiliation(s)
- Ksenia G. Kuznetsova
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Jakub Vašíček
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Dafni Skiadopoulou
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Janne Molnes
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Miriam Udler
- Department of Medicine, Massachusetts General Hospital, Boston, MA, United States of America
- Metabolism Program, Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
- Department of Medicine, Harvard Medical School, Boston, MA, United States of America
| | - Stefan Johansson
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Pål Rasmus Njølstad
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
| | - Alisa Manning
- Department of Medicine, Massachusetts General Hospital, Boston, MA, United States of America
- Metabolism Program, Broad Institute of MIT and Harvard, Cambridge, MA, United States of America
- Department of Medicine, Harvard Medical School, Boston, MA, United States of America
| | - Marc Vaudel
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
- Department of Genetics and Bioinformatics, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
15
|
Auble B, Dey J. Monogenetic Etiologies of Diabetes. Med Clin North Am 2024; 108:15-26. [PMID: 37951647 DOI: 10.1016/j.mcna.2023.05.013] [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] [Indexed: 11/14/2023]
Abstract
Maturity onset diabetes of the young (MODY) describes a group of non-autoimmune forms of diabetes that are characterized by mostly autosomal dominant, monogenic mutations resulting in decreased beta cell function in the pancreas. MODY accounts for roughly 1% to 5% of diabetes cases, and the optimal treatment for each MODY depends on the causative mutation. This article provides a review of MODY to aid providers with knowing what aspects of the history and physical exam should prompt further investigation for this group of conditions.
Collapse
Affiliation(s)
- Bethany Auble
- Medical College of Wisconsin, 9000 West Wisconsin Avenue, Milwaukee, WI 53226, USA.
| | - Justin Dey
- Medical College of Wisconsin Affiliated Hospitals, Inc., Graduate Medical Education, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
16
|
Mancera-Rincón P, Luna-España MC, Rincon O, Guzmán I, Alvarez M. Maturity-onset Diabetes of the Young Type 7 (MODY7) and the Krüppellike Factor 11 Mutation (KLF11). A Review. Curr Diabetes Rev 2024; 20:e210323214817. [PMID: 36944622 DOI: 10.2174/1573399819666230321114456] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 01/04/2023] [Accepted: 01/17/2023] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Maturity-onset diabetes of the young (MODY) is a rare disease due to a single gene mutation that affects several family members in most cases. The Krüppel-like factor 11 (KLF11) gene mutation is associated with decreased insulin sensitivity to high glucose levels. KLF 11 has been implicated in the pathogenesis of MODY type 7 but given its low prevalence, prolonged subclinical period, and the emergence of new information, doubts are raised about its association. METHODS A literature search of the PubMed, Scopus, and EBSCO databases was performed. The terms "Diabetes Mellitus, Type 2/genetics", "Mason-Type Diabetes" , "Maturity-Onset diabetes of the young", "KLF11 protein, human", and "Maturity-Onset Diabetes of the Young, Type 7" were used"., "Diagnosis" The search selection was not standardized. RESULTS The KLF1 mutation is rare and represents <1% of the mutations associated with monogenic diabetes. Its isolation in European family lines in the first studies and the emergence of new variants pose new diagnostic challenges. This article reviews the definition, epidemiology, pathophysiology, diagnosis, and treatment of MODY type 7. CONCLUSION MODY type 7 diabetes represents a rare form of monogenic diabetes with incomplete penetrance. Given its rarity, its association with impaired glucose metabolism has been questioned. Strict evaluation of glycemic control and the appearance of microvascular complications are key areas in the follow-up of patients diagnosed with MODY 7. More studies will be required to characterize the population with KLF11 mutation and clarify its correlation with MODY.
Collapse
Affiliation(s)
| | | | - Oswaldo Rincon
- Endocronology Department, Hospital Militar Central, Bogota, Colombia
| | - Issac Guzmán
- Endocronology Department, Hospital Militar Central, Bogota, Colombia
| | - Mauricio Alvarez
- Endocronology Department, Hospital Militar Central, Bogota, Colombia
| |
Collapse
|
17
|
Szczerbinski L, Florez JC. Precision medicine in diabetes - current trends and future directions. Is the future now? COMPREHENSIVE PRECISION MEDICINE 2024:458-483. [DOI: 10.1016/b978-0-12-824010-6.00021-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
18
|
Adadey SM, Mensah JA, Acquah KS, Abugri J, Osei-Yeboah R. Early-onset diabetes in Africa: A mini-review of the current genetic profile. Eur J Med Genet 2023; 66:104887. [PMID: 37995864 DOI: 10.1016/j.ejmg.2023.104887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023]
Abstract
Early-onset diabetes is poorly diagnosed partly due to its heterogeneity and variable presentations. Although several genes have been associated with the disease, these genes are not well studied in Africa. We sought to identify the major neonatal, early childhood, juvenile, or early-onset diabetes genes in Africa; and evaluate the available molecular methods used for investigating these gene variants. A literature search was conducted on PubMed, Scopus, Africa-Wide Information, and Web of Science databases. The retrieved records were screened and analyzed to identify genetic variants associated with early-onset diabetes. Although 319 records were retrieved, 32 were considered for the current review. Most of these records (22/32) were from North Africa. The disease condition was genetically heterogenous with most cases possessing unique gene variants. We identified 22 genes associated with early-onset diabetes, 9 of which had variants (n = 19) classified as pathogenic or likely pathogenic (PLP). Among the PLP variants, IER3IP1: p.(Leu78Pro) was the variant with the highest number of cases. There was limited data from West Africa, hence the contribution of genetic variability to early-onset diabetes in Africa could not be comprehensively evaluated. It is worth mentioning that most studies were focused on natural products as antidiabetics and only a few studies reported on the genetics of the disease. ABCC8 and KCNJ11 were implicated as major contributors to early-onset diabetes gene networks. Gene ontology analysis of the network associated ion channels, impaired glucose tolerance, and decreased insulin secretions to the disease. Our review highlights 9 genes from which PLP variants have been identified and can be considered for the development of an African diagnostic panel. There is a gap in early-onset diabetes genetic research from sub-Saharan Africa which is much needed to develop a comprehensive, efficient, and cost-effective genetic panel that will be useful in clinical practice on the continent and among the African diasporas.
Collapse
Affiliation(s)
- Samuel Mawuli Adadey
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana; School of Medicine and Health Science, University for Development Studies, Tamale, Ghana.
| | | | - Kojo Sekyi Acquah
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA.
| | - James Abugri
- Department of Biochemistry and Forensic Sciences, School of Chemical and Biochemical Sciences, C.K. Tedam University of Technology and Applied Sciences, Navrongo, Ghana.
| | - Richard Osei-Yeboah
- Centre for Global Health, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
19
|
Svalastoga P, Kaci A, Molnes J, Solheim MH, Johansson BB, Krogvold L, Skrivarhaug T, Valen E, Johansson S, Molven A, Sagen JV, Søfteland E, Bjørkhaug L, Tjora E, Aukrust I, Njølstad PR. Characterisation of HNF1A variants in paediatric diabetes in Norway using functional and clinical investigations to unmask phenotype and monogenic diabetes. Diabetologia 2023; 66:2226-2237. [PMID: 37798422 PMCID: PMC10627920 DOI: 10.1007/s00125-023-06012-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 06/09/2023] [Indexed: 10/07/2023]
Abstract
AIMS/HYPOTHESIS Correctly diagnosing MODY is important, as individuals with this diagnosis can discontinue insulin injections; however, many people are misdiagnosed. We aimed to develop a robust approach for determining the pathogenicity of variants of uncertain significance in hepatocyte nuclear factor-1 alpha (HNF1A)-MODY and to obtain an accurate estimate of the prevalence of HNF1A-MODY in paediatric cases of diabetes. METHODS We extended our previous screening of the Norwegian Childhood Diabetes Registry by 830 additional samples and comprehensively genotyped HNF1A variants in autoantibody-negative participants using next-generation sequencing. Carriers of pathogenic variants were treated by local healthcare providers, and participants with novel likely pathogenic variants and variants of uncertain significance were enrolled in an investigator-initiated, non-randomised, open-label pilot study (ClinicalTrials.gov registration no. NCT04239586). To identify variants associated with HNF1A-MODY, we functionally characterised their pathogenicity and assessed the carriers' phenotype and treatment response to sulfonylurea. RESULTS In total, 615 autoantibody-negative participants among 4712 cases of paediatric diabetes underwent genetic sequencing, revealing 19 with HNF1A variants. We identified nine carriers with novel variants classified as variants of uncertain significance or likely to be pathogenic, while the remaining ten participants carried five pathogenic variants previously reported. Of the nine carriers with novel variants, six responded favourably to sulfonylurea. Functional investigations revealed their variants to be dysfunctional and demonstrated a correlation with the resulting phenotype, providing evidence for reclassifying these variants as pathogenic. CONCLUSIONS/INTERPRETATION Based on this robust classification, we estimate that the prevalence of HNF1A-MODY is 0.3% in paediatric diabetes. Clinical phenotyping is challenging and functional investigations provide a strong complementary line of evidence. We demonstrate here that combining clinical phenotyping with functional protein studies provides a powerful tool to obtain a precise diagnosis of HNF1A-MODY.
Collapse
Affiliation(s)
- Pernille Svalastoga
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Alba Kaci
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Center for Laboratory Medicine, Østfold Hospital Trust, Grålum, Norway
| | - Janne Molnes
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Marie H Solheim
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bente B Johansson
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lars Krogvold
- Division of Childhood and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Torild Skrivarhaug
- Division of Childhood and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Eivind Valen
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
- Sars International Centre for Marine Molecular Biology, University of Bergen, Bergen, Norway
| | - Stefan Johansson
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Anders Molven
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Jørn V Sagen
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Eirik Søfteland
- Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Lise Bjørkhaug
- Department of Safety, Chemistry, and Biomedical Laboratory Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | - Erling Tjora
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
| | - Ingvild Aukrust
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Pål R Njølstad
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway.
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
20
|
Wu HX, Chu TY, Iqbal J, Jiang HL, Li L, Wu YX, Zhou HD. Cardio-cerebrovascular Outcomes in MODY, Type 1 Diabetes, and Type 2 Diabetes: A Prospective Cohort Study. J Clin Endocrinol Metab 2023; 108:2970-2980. [PMID: 37093977 DOI: 10.1210/clinem/dgad233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/09/2023] [Accepted: 04/20/2023] [Indexed: 04/26/2023]
Abstract
CONTEXT Cardio-cerebrovascular events are severe complications of diabetes. OBJECTIVE We aim to compare the incident risk of cardio-cerebrovascular events in maturity onset diabetes of the young (MODY), type 1 diabetes, and type 2 diabetes. METHODS Type 1 diabetes, type 2 diabetes, and MODY were diagnosed by whole exome sequencing. The primary endpoint was the occurrence of the first major adverse cardiovascular event (MACE), including acute myocardial infarction, heart failure, stroke, unstable angina pectoris, and cardio-cerebrovascular-related mortality. Cox proportional hazards models were applied and adjusted to calculate hazard ratios (HRs) and 95% CIs for the incident risk of MACE in type 1 diabetes, type 2 diabetes, MODY, and MODY subgroups compared with people without diabetes (control group). RESULTS Type 1 diabetes, type 2 diabetes, and MODY accounted for 2.7%, 68.1%, and 11.4% of 26 198 participants with diabetes from UK Biobank. During a median follow-up of 13 years, 1028 MACEs occurred in the control group, contrasting with 70 events in patients with type 1 diabetes (HR 2.15, 95% CI 1.69-2.74, P < .05), 5020 events in patients with type 2 diabetes (HR 7.02, 95% CI 6.56-7.51, P < .05), and 717 events in MODY (HR 5.79, 95% CI 5.26-6.37, P < .05). The hazard of MACE in HNF1B-MODY was highest among MODY subgroups (HR 11.00, 95% CI 5.47-22.00, P = 1.5 × 10-11). CONCLUSION MODY diagnosed by genetic analysis represents higher prevalence than the clinical diagnosis in UK Biobank. The risk of incident cardio-cerebrovascular events in MODY ranks between type 1 diabetes and type 2 diabetes.
Collapse
Affiliation(s)
- Hui-Xuan Wu
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Key Laboratory of Diabetes Immunology Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Tian-Yao Chu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 41000, Hunan, China
| | - Junaid Iqbal
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Key Laboratory of Diabetes Immunology Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Hong-Li Jiang
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Key Laboratory of Diabetes Immunology Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Long Li
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Key Laboratory of Diabetes Immunology Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yan-Xuan Wu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 15000, China
| | - Hou-De Zhou
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory for Metabolic Bone Diseases, Key Laboratory of Diabetes Immunology Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| |
Collapse
|
21
|
Murphy R, Colclough K, Pollin TI, Ikle JM, Svalastoga P, Maloney KA, Saint-Martin C, Molnes J, Misra S, Aukrust I, de Franco E, Flanagan SE, Njølstad PR, Billings LK, Owen KR, Gloyn AL. The use of precision diagnostics for monogenic diabetes: a systematic review and expert opinion. COMMUNICATIONS MEDICINE 2023; 3:136. [PMID: 37794142 PMCID: PMC10550998 DOI: 10.1038/s43856-023-00369-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/21/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Monogenic diabetes presents opportunities for precision medicine but is underdiagnosed. This review systematically assessed the evidence for (1) clinical criteria and (2) methods for genetic testing for monogenic diabetes, summarized resources for (3) considering a gene or (4) variant as causal for monogenic diabetes, provided expert recommendations for (5) reporting of results; and reviewed (6) next steps after monogenic diabetes diagnosis and (7) challenges in precision medicine field. METHODS Pubmed and Embase databases were searched (1990-2022) using inclusion/exclusion criteria for studies that sequenced one or more monogenic diabetes genes in at least 100 probands (Question 1), evaluated a non-obsolete genetic testing method to diagnose monogenic diabetes (Question 2). The risk of bias was assessed using the revised QUADAS-2 tool. Existing guidelines were summarized for questions 3-5, and review of studies for questions 6-7, supplemented by expert recommendations. Results were summarized in tables and informed recommendations for clinical practice. RESULTS There are 100, 32, 36, and 14 studies included for questions 1, 2, 6, and 7 respectively. On this basis, four recommendations for who to test and five on how to test for monogenic diabetes are provided. Existing guidelines for variant curation and gene-disease validity curation are summarized. Reporting by gene names is recommended as an alternative to the term MODY. Key steps after making a genetic diagnosis and major gaps in our current knowledge are highlighted. CONCLUSIONS We provide a synthesis of current evidence and expert opinion on how to use precision diagnostics to identify individuals with monogenic diabetes.
Collapse
Affiliation(s)
- Rinki Murphy
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
- Auckland Diabetes Centre, Te Whatu Ora Health New Zealand, Te Tokai Tumai, Auckland, New Zealand.
| | - Kevin Colclough
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom
| | - Toni I Pollin
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jennifer M Ikle
- Department of Pediatrics, Division of Endocrinology & Diabetes, Stanford School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford School of Medicine, Stanford, CA, USA
| | - Pernille Svalastoga
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kristin A Maloney
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cécile Saint-Martin
- Department of Medical Genetics, AP-HP Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Janne Molnes
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Shivani Misra
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Ingvild Aukrust
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Elisa de Franco
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Sarah E Flanagan
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Pål R Njølstad
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Liana K Billings
- Division of Endocrinology, NorthShore University HealthSystem, Skokie, IL, USA
- Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Katharine R Owen
- Oxford Center for Diabetes, Endocrinology & Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Anna L Gloyn
- Department of Pediatrics, Division of Endocrinology & Diabetes, Stanford School of Medicine, Stanford, CA, USA.
- Stanford Diabetes Research Center, Stanford School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
22
|
Lau HH, Krentz NAJ, Abaitua F, Perez-Alcantara M, Chan JW, Ajeian J, Ghosh S, Lee Y, Yang J, Thaman S, Champon B, Sun H, Jha A, Hoon S, Tan NS, Gardner DSL, Kao SL, Tai ES, Gloyn AL, Teo AKK. PAX4 loss of function increases diabetes risk by altering human pancreatic endocrine cell development. Nat Commun 2023; 14:6119. [PMID: 37777536 PMCID: PMC10542369 DOI: 10.1038/s41467-023-41860-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 09/20/2023] [Indexed: 10/02/2023] Open
Abstract
The coding variant (p.Arg192His) in the transcription factor PAX4 is associated with an altered risk for type 2 diabetes (T2D) in East Asian populations. In mice, Pax4 is essential for beta cell formation but its role on human beta cell development and/or function is unknown. Participants carrying the PAX4 p.His192 allele exhibited decreased pancreatic beta cell function compared to homozygotes for the p.192Arg allele in a cross-sectional study in which we carried out an intravenous glucose tolerance test and an oral glucose tolerance test. In a pedigree of a patient with young onset diabetes, several members carry a newly identified p.Tyr186X allele. In the human beta cell model, EndoC-βH1, PAX4 knockdown led to impaired insulin secretion, reduced total insulin content, and altered hormone gene expression. Deletion of PAX4 in human induced pluripotent stem cell (hiPSC)-derived islet-like cells resulted in derepression of alpha cell gene expression. In vitro differentiation of hiPSCs carrying PAX4 p.His192 and p.X186 risk alleles exhibited increased polyhormonal endocrine cell formation and reduced insulin content that can be reversed with gene correction. Together, we demonstrate the role of PAX4 in human endocrine cell development, beta cell function, and its contribution to T2D-risk.
Collapse
Affiliation(s)
- Hwee Hui Lau
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Proteos, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Nicole A J Krentz
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Fernando Abaitua
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | - Jun-Wei Chan
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Proteos, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Jila Ajeian
- Oxford Centre for Diabetes Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Soumita Ghosh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Yunkyeong Lee
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jing Yang
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Swaraj Thaman
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Benoite Champon
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Han Sun
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alokkumar Jha
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Shawn Hoon
- Molecular Engineering Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Proteos, Singapore
| | - Nguan Soon Tan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | | | - Shih Ling Kao
- Department of Medicine, National University Hospital and National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - E Shyong Tai
- Department of Medicine, National University Hospital and National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Anna L Gloyn
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA.
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Proteos, Singapore.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
23
|
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
|
24
|
Hattersley AT. Laboratory Guidelines Are Needed for Diagnostic Genetic Testing for Monogenic Diabetes. Clin Chem 2023:hvad093. [PMID: 37473454 DOI: 10.1093/clinchem/hvad093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 07/22/2023]
Affiliation(s)
- Andrew T Hattersley
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
25
|
Samadli S, Zhou Q, Zheng B, Gu W, Zhang A. From glucose sensing to exocytosis: takes from maturity onset diabetes of the young. Front Endocrinol (Lausanne) 2023; 14:1188301. [PMID: 37255971 PMCID: PMC10226665 DOI: 10.3389/fendo.2023.1188301] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/28/2023] [Indexed: 06/01/2023] Open
Abstract
Monogenic diabetes gave us simplified models of complex molecular processes occurring within β-cells, which allowed to explore the roles of numerous proteins from single protein perspective. Constellation of characteristic phenotypic features and wide application of genetic sequencing techniques to clinical practice, made the major form of monogenic diabetes - the Maturity Onset Diabetes of the Young to be distinguishable from type 1, type 2 as well as neonatal diabetes mellitus and understanding underlying molecular events for each type of MODY contributed to the advancements of antidiabetic therapy and stem cell research tremendously. The functional analysis of MODY-causing proteins in diabetes development, not only provided better care for patients suffering from diabetes, but also enriched our comprehension regarding the universal cellular processes including transcriptional and translational regulation, behavior of ion channels and transporters, cargo trafficking, exocytosis. In this review, we will overview structure and function of MODY-causing proteins, alterations in a particular protein arising from the deleterious mutations to the corresponding gene and their consequences, and translation of this knowledge into new treatment strategies.
Collapse
Affiliation(s)
- Sama Samadli
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Department of Pediatric Diseases II, Azerbaijan Medical University, Baku, Azerbaijan
| | - Qiaoli Zhou
- Department of Endocrinology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Bixia Zheng
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Gu
- Department of Endocrinology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
Murphy R, Colclough K, Pollin TI, Ikle JM, Svalastoga P, Maloney KA, Saint-Martin C, Molnes J, Misra S, Aukrust I, de Franco A, Flanagan SE, Njølstad PR, Billings LK, Owen KR, Gloyn AL. A Systematic Review of the use of Precision Diagnostics in Monogenic Diabetes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.15.23288269. [PMID: 37131594 PMCID: PMC10153302 DOI: 10.1101/2023.04.15.23288269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Monogenic forms of diabetes present opportunities for precision medicine as identification of the underlying genetic cause has implications for treatment and prognosis. However, genetic testing remains inconsistent across countries and health providers, often resulting in both missed diagnosis and misclassification of diabetes type. One of the barriers to deploying genetic testing is uncertainty over whom to test as the clinical features for monogenic diabetes overlap with those for both type 1 and type 2 diabetes. In this review, we perform a systematic evaluation of the evidence for the clinical and biochemical criteria used to guide selection of individuals with diabetes for genetic testing and review the evidence for the optimal methods for variant detection in genes involved in monogenic diabetes. In parallel we revisit the current clinical guidelines for genetic testing for monogenic diabetes and provide expert opinion on the interpretation and reporting of genetic tests. We provide a series of recommendations for the field informed by our systematic review, synthesizing evidence, and expert opinion. Finally, we identify major challenges for the field and highlight areas for future research and investment to support wider implementation of precision diagnostics for monogenic diabetes.
Collapse
Affiliation(s)
- Rinki Murphy
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Auckland Diabetes Centre, Te Whatu Ora Health New Zealand, Te Tokai Tumai, Auckland, New Zealand
| | - Kevin Colclough
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom
| | - Toni I Pollin
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jennifer M Ikle
- Department of Pediatrics, Division of Endocrinology & Diabetes, Stanford School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford School of Medicine, Stanford, CA, USA
| | - Pernille Svalastoga
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kristin A Maloney
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cécile Saint-Martin
- Department of Medical Genetics, AP-HP Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Janne Molnes
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Shivani Misra
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Ingvild Aukrust
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - aiElisa de Franco
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sarah E Flanagan
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, UK
| | - Pål R Njølstad
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Liana K Billings
- Division of Endocrinology, NorthShore University HealthSystem, Skokie, IL, USA; Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Katharine R Owen
- Oxford Center for Diabetes, Endocrinology & Metabolism, University of Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Anna L Gloyn
- Department of Pediatrics, Division of Endocrinology & Diabetes, Stanford School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| |
Collapse
|
27
|
Harsunen M, Kettunen JLT, Härkönen T, Dwivedi O, Lehtovirta M, Vähäsalo P, Veijola R, Ilonen J, Miettinen PJ, Knip M, Tuomi T. Identification of monogenic variants in more than ten per cent of children without type 1 diabetes-related autoantibodies at diagnosis in the Finnish Pediatric Diabetes Register. Diabetologia 2023; 66:438-449. [PMID: 36418577 PMCID: PMC9892083 DOI: 10.1007/s00125-022-05834-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/05/2022] [Indexed: 11/25/2022]
Abstract
AIMS/HYPOTHESIS Monogenic forms of diabetes (MODY, neonatal diabetes mellitus and syndromic forms) are rare, and affected individuals may be misclassified and treated suboptimally. The prevalence of type 1 diabetes is high in Finnish children but systematic screening for monogenic diabetes has not been conducted. We assessed the prevalence and clinical manifestations of monogenic diabetes in children initially registered with type 1 diabetes in the Finnish Pediatric Diabetes Register (FPDR) but who had no type 1 diabetes-related autoantibodies (AABs) or had only low-titre islet cell autoantibodies (ICAs) at diagnosis. METHODS The FPDR, covering approximately 90% of newly diagnosed diabetic individuals aged ≤15 years in Finland starting from 2002, includes data on diabetes-associated HLA genotypes and AAB data (ICA, and autoantibodies against insulin, GAD, islet antigen 2 and zinc transporter 8) at diagnosis. A next generation sequencing gene panel including 42 genes was used to identify monogenic diabetes. We interpreted the variants in HNF1A by using the gene-specific standardised criteria and reported pathogenic and likely pathogenic findings only. For other genes, we also reported variants of unknown significance if an individual's phenotype suggested monogenic diabetes. RESULTS Out of 6482 participants, we sequenced DNA for 152 (2.3%) testing negative for all AABs and 49 (0.8%) positive only for low-titre ICAs (ICAlow). A monogenic form of diabetes was revealed in 19 (12.5%) of the AAB-negative patients (14 [9.2%] had pathogenic or likely pathogenic variants) and two (4.1%) of the ICAlow group. None had ketoacidosis at diagnosis or carried HLA genotypes conferring high risk for type 1 diabetes. The affected genes were GCK, HNF1A, HNF4A, HNF1B, INS, KCNJ11, RFX6, LMNA and WFS1. A switch from insulin to oral medication was successful in four of five patients with variants in HNF1A, HNF4A or KCNJ11. CONCLUSIONS/INTERPRETATION More than 10% of AAB-negative children with newly diagnosed diabetes had a genetic finding associated with monogenic diabetes. Because the genetic diagnosis can lead to major changes in treatment, we recommend referring all AAB-negative paediatric patients with diabetes for genetic testing. Low-titre ICAs in the absence of other AABs does not always indicate a diagnosis of type 1 diabetes.
Collapse
Affiliation(s)
- Minna Harsunen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland.
| | - Jarno L T Kettunen
- Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland.
- Abdominal Centre, Endocrinology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Finnish Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland.
| | - Taina Härkönen
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
| | - Om Dwivedi
- Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Finnish Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko Lehtovirta
- Finnish Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - Paula Vähäsalo
- Department of Pediatrics, PEDEGO Research Unit, University of Oulu, Oulu, Finland
- Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
- Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Riitta Veijola
- Department of Pediatrics, PEDEGO Research Unit, University of Oulu, Oulu, Finland
- Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
- Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Päivi J Miettinen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Translational Stem Cell Biology and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikael Knip
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland
| | - Tiinamaija Tuomi
- Folkhälsan Research Center, Biomedicum Helsinki, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
- Abdominal Centre, Endocrinology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Finnish Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
28
|
Guan G, Qin T, Zhao LL, Jin P. Genetic and Functional Analyses of the Novel KLF11 Pro193Thr Variant in a Three-Generation Family with MODY7. Horm Metab Res 2023; 55:136-141. [PMID: 36241199 DOI: 10.1055/a-1961-6281] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
KLF11 regulates insulin gene expression through binding to the insulin promoter and has been reported as a causative gene for maturity-onset diabetes of the young 7 (MODY7). Here, we report a novel KLF11 variant associated with a three-generation family with early childhood-onset diabetes and explore its clinical and functional characteristics. The three-generational pedigree contains five patients affected by diabetes. The pathogenic variant identified by whole-exome sequencing was further confirmed by Sanger sequencing and pedigree verification. Luciferase reporter assays and glucose-stimulated insulin secretion were used to examine whether the KLF11 variant binds to the insulin promoter and regulate insulin secretion in vitro. The proband, his son, and his uncle exhibited hyperglycemia at ages 32, 13 and 71 years, respectively. All three patients showed characteristics of metabolic syndrome (obesity, dyslipidemia, and diabetes), but the insulin secretion of islet β-cells was impaired. A novel heterozygous missense variant, c.577 C>A (p.Pro193Thr) of the KLF11 gene was detected in all three patients. This variant co-segregates with the diabetes phenotype, consistent with an autosomal dominant disorder. The identified KLF11 p.Pro193Thr variant drastically decreased the transcriptional activity of KLF11, as demonstrated by luciferase reporter assay. Functional analyses revealed that the KLF11 Pro193Thr variant inhibited glucose-stimulated insulin secretion. We identified a novel KLF11 Pro193Thr variant in a three generation family with MODY7. These findings shed light on the molecular mechanisms underlying the pathogenesis of MODY7 and expand the genotype and clinical spectrum of MODY7.
Collapse
Affiliation(s)
- Gaopeng Guan
- Department of Endocrinology, Central South University Third Xiangya Hospital, Changsha, China
| | - Tiantian Qin
- Department of Endocrinology, Central South University Third Xiangya Hospital, Changsha, China
| | - Li-Ling Zhao
- Department of Endocrinology, Central South University Third Xiangya Hospital, Changsha, China
| | - Ping Jin
- Department of Endocrinology, Central South University Third Xiangya Hospital, Changsha, China
| |
Collapse
|
29
|
Santos Monteiro S, da Silva Santos T, Fonseca L, Assunção G, Lopes AM, Duarte DB, Soares AR, Laranjeira F, Ribeiro I, Pinto E, Rocha S, Barbosa Gouveia S, Vazquez-Mosquera ME, Oliveira MJ, Borges T, Cardoso MH. Maturity-onset diabetes of the young in a large Portuguese cohort. Acta Diabetol 2023; 60:83-91. [PMID: 36208343 DOI: 10.1007/s00592-022-01980-2] [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] [Received: 07/25/2022] [Accepted: 09/22/2022] [Indexed: 01/07/2023]
Abstract
AIMS Monogenic forms of diabetes that develop with autosomal dominant inheritance are classically aggregated in the Maturity-Onset Diabetes of the Young (MODY) categories. Despite increasing awareness, its true prevalence remains largely underestimated. We describe a Portuguese cohort of individuals with suspected monogenic diabetes who were genetically evaluated for MODY-causing genes. METHODS This single-center retrospective cohort study enrolled patients with positive genetic testing for MODY between 2015 and 2021. Automatic sequencing and, in case of initial negative results, next-generation sequencing were performed. Their clinical and molecular characteristics were described. RESULTS Eighty individuals were included, 55 with likely pathogenic/pathogenic variants in one of the MODY genes and 25 MODY-positive family members, identified by cascade genetic testing. The median age at diabetes diagnosis was 23 years, with a median HbA1c of 6.5%. The most frequently mutated genes were identified in HNF1A (40%), GCK (34%) and HNF4A (13%), followed by PDX1, HNF1B, INS, KCNJ11 and APPL1. Thirty-six unique variants were found (29 missense and 7 frameshift variants), of which ten (28%) were novel. CONCLUSIONS Our data highlights the importance of genetic testing in the diagnosis of MODY and the establishment of its subtypes, leading to more personalized treatment and follow-up strategies.
Collapse
Affiliation(s)
- Sílvia Santos Monteiro
- Division of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário do Porto, Largo Professor Abel Salazar, 4099-001, Porto, Portugal.
| | - Tiago da Silva Santos
- Division of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário do Porto, Largo Professor Abel Salazar, 4099-001, Porto, Portugal
| | - Liliana Fonseca
- Division of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário do Porto, Largo Professor Abel Salazar, 4099-001, Porto, Portugal
| | - Guilherme Assunção
- Division of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário do Porto, Largo Professor Abel Salazar, 4099-001, Porto, Portugal
| | - Ana M Lopes
- Division of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário do Porto, Largo Professor Abel Salazar, 4099-001, Porto, Portugal
| | - Diana B Duarte
- Division of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário do Porto, Largo Professor Abel Salazar, 4099-001, Porto, Portugal
| | - Ana Rita Soares
- Division of Medical Genetics, Centro de Genética Médica Doutor Jacinto Magalhães, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Francisco Laranjeira
- Division of Genetic Biochemistry. Centro de Genética Médica Doutor Jacinto Magalhães, Centro Hospitalar Universitário do Porto, Porto, Portugal
- Unit for Multidisciplinar Biomedical Research (UMIB), Instituto de Ciências Biomédicas Abel Salazar. Universidade do Porto, Porto, Portugal
| | - Isaura Ribeiro
- Division of Genetic Biochemistry. Centro de Genética Médica Doutor Jacinto Magalhães, Centro Hospitalar Universitário do Porto, Porto, Portugal
- Unit for Multidisciplinar Biomedical Research (UMIB), Instituto de Ciências Biomédicas Abel Salazar. Universidade do Porto, Porto, Portugal
| | - Eugénia Pinto
- Division of Genetic Biochemistry. Centro de Genética Médica Doutor Jacinto Magalhães, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Sónia Rocha
- Division of Genetic Biochemistry. Centro de Genética Médica Doutor Jacinto Magalhães, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Sofia Barbosa Gouveia
- University Clinical Hospital of Santiago de Compostela. IDIS, CIBERER, MetabERN, 15701, Santiago de Compostela, Spain
| | | | - Maria João Oliveira
- Division of Pediatric Endocrinology. Department of Pediatrics. Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Teresa Borges
- Division of Pediatric Endocrinology. Department of Pediatrics. Centro Materno-Infantil do Norte, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Maria Helena Cardoso
- Division of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário do Porto, Largo Professor Abel Salazar, 4099-001, Porto, Portugal
| |
Collapse
|
30
|
Cao B, Liu M, Zhang Y, Chen J, Li X, Su C, Yang W, Liu M, Wu D, Li W, Liang X, Wang Q, Wei H, Gong C. An effective preselection criterion for MODY with an increasingly positive genetic testing rate by NGS: results from two cohorts of Chinese children. Am J Physiol Endocrinol Metab 2022; 323:E529-E534. [PMID: 36383636 DOI: 10.1152/ajpendo.00171.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The purpose of this study was to determine the frequency of maturity-onset diabetes of the young (MODY) in two selected cohorts of Chinese children with diabetes and clinically suspected MODY, using next-generation sequencing (NGS). Ninety-three children who met the comprehensive criteria of suspected MODY were enrolled in two cohorts. A custom NGS panel or a whole exon group was used for sequencing. We identified 55/93 (59.1%) children with pathogenic and likely pathogenic MODY variants. Forty-two (76.3%) were confirmed to have the GCK (MODY2) mutation. Additionally, five had the HNF1A (MODY3), two the HNF1B (MODY5), one the 17q12 microdeletion (MODY5), two the HNF4A (MODY1), two the ABCC8 (MODY12), and one the PDX1 mutation (MODY4). Of these, 13 novel variants were detected in different genes. By comparing the gene-positive with gene-negative children, we found that discriminatory factors for MODY at diagnosis included lower HbA1c [7.4% vs. 10.2% (53 vs. 86 mmol/mol); P = 0.002], lower body mass index z score (0.2 vs. 1.0; P = 0.01), lower onset age (8.1 vs. 11.2 years; P = 0.001), and lower C-peptide (1.4 vs. 2.5 ng/mL; P = 0.02). In conclusion, the criteria used in this study for screening MODY are effective, and MODY2 is the most common subtype (76%), followed by MODY3 and MODY5. Some rare MODY subtypes have been reported in Chinese children.NEW & NOTEWORTHY We proved the clinical suspicion of maturity-onset diabetes of the young (MODY) according to the comprehensive criterion for next-generation sequencing testing, which helps to identify both common and rare MODYs, leading to accurate diagnosis and personalized treatment.
Collapse
Affiliation(s)
- Bingyan Cao
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Meijuan Liu
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yingxian Zhang
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Jiajia Chen
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xiaoqiao Li
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Chang Su
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wei Yang
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Min Liu
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Di Wu
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wenjing Li
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xuejun Liang
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Qiao Wang
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Haiyan Wei
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Chunxiu Gong
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
31
|
Billings LK, Shi Z, Resurreccion WK, Wang C, Wei J, Pollin TI, Udler MS, Xu J. Statistical evidence for high-penetrance MODY-causing genes in a large population-based cohort. Endocrinol Diabetes Metab 2022; 5:e372. [PMID: 36208030 PMCID: PMC9659663 DOI: 10.1002/edm2.372] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/29/2022] [Accepted: 09/04/2022] [Indexed: 11/15/2022] Open
Abstract
AIMS Numerous genes have been proposed as causal for maturity-onset diabetes of the young (MODY). Scoring systems to annotate mutation pathogenicity have been widely used; however, statistical evidence for being a highly penetrant MODY gene has not been well-established. METHODS Participants were from the UK Biobank with whole-exome sequencing data, including 14,622 with and 185,509 without diagnosis of diabetes. Pathogenic/likely pathogenic (P/LP) mutations in 14 reported and 3 possible MODY genes were annotated using American College of Medical Genetics criteria. Evidence for being a high-penetrant MODY gene used two statistical criteria: frequency of aggregate P/LP mutations in each gene are (1) significantly more common in participants with a diagnosis of diabetes than without using the SKAT-O (p < .05) and (2) lower than the maximum credible frequency in the general population. RESULTS Among the 17 genes, 6 (GCK, HNF1A, HNF4A, NEUROD1, KCNJ11 and HNF1B) met both criteria, 7 (ABCC8, KLF11, RFX6, PCBD1, WFS1, INS and PDX1) met only one criterion, and the remaining 4 (CEL, BLK, APPL1 and PAX4) failed both criteria, and were classified as 'consistent', 'inconclusive' and 'inconsistent' for being highly penetrant diabetes genes, respectively. Diabetes participants with mutations in the 'consistent' genes had clinical presentations that were most consistent with MODY. In contrast, the 'inconclusive' and 'inconsistent' genes did not differ clinically from non-carriers in diabetes-related characteristics. CONCLUSIONS Data from a large population-based study provided novel statistical evidence to identify 6 MODY genes as consistent with being highly penetrant. These results have potential implications for interpreting genetic testing results and clinical diagnosis of MODY.
Collapse
Affiliation(s)
- Liana K. Billings
- Department of MedicineNorthShore University HealthSystemSkokieIllinoisUSA
- University of Chicago Pritzker School of MedicineChicagoIllinoisUSA
| | - Zhuqing Shi
- Program for Personalized Cancer CareNorthShore University HealthSystemEvanstonIllinoisUSA
| | - W. Kyle Resurreccion
- Program for Personalized Cancer CareNorthShore University HealthSystemEvanstonIllinoisUSA
| | - Chi‐Hsiung Wang
- Program for Personalized Cancer CareNorthShore University HealthSystemEvanstonIllinoisUSA
| | - Jun Wei
- Program for Personalized Cancer CareNorthShore University HealthSystemEvanstonIllinoisUSA
| | - Toni I. Pollin
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, and Program in Personalized and Genomic MedicineUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Miriam S. Udler
- Diabetes UnitMassachusetts General HospitalBostonMassachusettsUSA
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Jianfeng Xu
- University of Chicago Pritzker School of MedicineChicagoIllinoisUSA
- Program for Personalized Cancer CareNorthShore University HealthSystemEvanstonIllinoisUSA
| |
Collapse
|
32
|
Younis H, Ha SE, Jorgensen BG, Verma A, Ro S. Maturity-Onset Diabetes of the Young: Mutations, Physiological Consequences, and Treatment Options. J Pers Med 2022; 12:1762. [PMID: 36573710 PMCID: PMC9697644 DOI: 10.3390/jpm12111762] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 02/01/2023] Open
Abstract
Maturity-Onset Diabetes of the Young (MODY) is a rare form of diabetes which affects between 1% and 5% of diagnosed diabetes cases. Clinical characterizations of MODY include onset of diabetes at an early age (before the age of 30), autosomal dominant inheritance pattern, impaired glucose-induced secretion of insulin, and hyperglycemia. Presently, 14 MODY subtypes have been identified. Within these subtypes are several mutations which contribute to the different MODY phenotypes. Despite the identification of these 14 subtypes, MODY is often misdiagnosed as type 1 or type 2 diabetes mellitus due to an overlap in clinical features, high cost and limited availability of genetic testing, and unfamiliarity with MODY outside of the medical profession. The primary aim of this review is to investigate the genetic characterization of the MODY subtypes. Additionally, this review will elucidate the link between the genetics, function, and clinical manifestations of MODY in each of the 14 subtypes. In providing this knowledge, we hope to assist in the accurate diagnosis of MODY patients and, subsequently, in ensuring they receive appropriate treatment.
Collapse
Affiliation(s)
- Hazar Younis
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Se Eun Ha
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Brian G. Jorgensen
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Arushi Verma
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
- RosVivo Therapeutics, Applied Research Facility, Reno, NV 89557, USA
| |
Collapse
|
33
|
Srinivasan S, Todd J. The Genetics of Type 2 Diabetes in Youth: Where We Are and the Road Ahead. J Pediatr 2022; 247:17-21. [PMID: 35660490 PMCID: PMC9833991 DOI: 10.1016/j.jpeds.2022.05.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 01/13/2023]
Affiliation(s)
- Shylaja Srinivasan
- Department of Pediatrics, University of California San Francisco, San Francisco, CA.
| | - Jennifer Todd
- Department of Pediatrics, University of Vermont, Burlington, VT
| |
Collapse
|
34
|
Colclough K, Ellard S, Hattersley A, Patel K. Syndromic Monogenic Diabetes Genes Should Be Tested in Patients With a Clinical Suspicion of Maturity-Onset Diabetes of the Young. Diabetes 2022; 71:530-537. [PMID: 34789499 PMCID: PMC7612420 DOI: 10.2337/db21-0517] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022]
Abstract
At present, outside of infancy, genetic testing for monogenic diabetes is typically for mutations in maturity-onset diabetes of the young (MODY) genes that predominantly result in isolated diabetes. Monogenic diabetes syndromes are usually only tested for when supported by specific syndromic clinical features. How frequently patients with suspected MODY have a mutation in a monogenic syndromic diabetes gene is unknown and thus missed by present testing regimes. We performed genetic testing of 27 monogenic diabetes genes (including 18 associated with syndromic diabetes) for 1,280 patients with a clinical suspicion of MODY who were not suspected of having monogenic syndromic diabetes. We confirmed monogenic diabetes in 297 (23%) patients. Mutations in seven different syndromic diabetes genes accounted for 19% (95% CI 15-24%) of all monogenic diabetes. The mitochondrial m.3243A>G and mutations in HNF1B were responsible for the majority of mutations in syndromic diabetes genes. They were also the 4th and 5th most common causes of monogenic diabetes overall. These patients lacked typical features, and their diabetes phenotypes overlapped with patients with nonsyndromic monogenic diabetes. Syndromic monogenic diabetes genes (particularly m.3243A>G and HNF1B) should be routinely tested in patients with suspected MODY who do not have typical features of a genetic syndrome.
Collapse
Affiliation(s)
- Kevin Colclough
- Exeter Genomics Laboratory, Royal Devon and Exeter NHS Foundation Trust, Exeter, U.K
| | - Sian Ellard
- Exeter Genomics Laboratory, Royal Devon and Exeter NHS Foundation Trust, Exeter, U.K
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Andrew Hattersley
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Kashyap Patel
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| |
Collapse
|