1
|
Astudillo MF, Winter WE, Billings LK, Kreienkamp R, Balasubramanyam A, Redondo MJ, Tosur M. Identification of atypical pediatric diabetes mellitus cases using electronic medical records. BMJ Open Diabetes Res Care 2024; 12:e004471. [PMID: 39510599 PMCID: PMC11551976 DOI: 10.1136/bmjdrc-2024-004471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/04/2024] [Indexed: 11/15/2024] Open
Abstract
INTRODUCTION There are no established methods to identify children with atypical diabetes for further study. We aimed to develop strategies to systematically ascertain cases of atypical pediatric diabetes using electronic medical records (EMR). RESEARCH DESIGN AND METHODS We tested two strategies in a large pediatric hospital in the USA. Strategy 1: we designed a questionnaire to rule out typical diabetes and applied it to the EMR of 100 youth with diabetes. Strategy 2: we built three electronic queries to generate reports of three atypical pediatric diabetes phenotypes: unknown type, type 2 diabetes (T2D) diagnosed <10 years old and autoantibody-negative type 1 diabetes (AbNegT1D). RESULTS Strategy 1 identified six cases (6%) of atypical diabetes (mean diagnosis age=11±2.6 years, 16.6% men, 33% non-Hispanic white (NHW) and 66.6% Hispanic). Strategy 2: unknown diabetes type: n=68 (1%) out of 6676 patients with diabetes; mean diagnosis age=12.6±3.3 years, 32.8% men, 23.8% NHW, 47.6% Hispanic, 25.4% African American (AA), 3.2% other. T2D <10 years old: n=64 (6.6%) out of 1142 patients with T2D; mean diagnosis age=8.6±1.6 years, 20.3% men, 4.7% NHW, 65.6% Hispanic, 28.1% AA, 1.6% other. AbNegT1D: n=38 (5.6%) out of 680 patients with new onset T1D; mean diagnosis age=11.3±3.8 years; 57.9% men, 50% NHW, 19.4% Hispanic, 22.3% AA, 8.3% other. CONCLUSIONS In sum, we identified 1%-6.6% of atypical diabetes cases in a pediatric diabetes population with high racial and ethnic diversity using systematic review of the EMR. Better identification of these cases using unbiased approaches may advance precision diabetes.
Collapse
Affiliation(s)
- Marcela F Astudillo
- Texas Children's Hospital—Pediatric Diabetes & Endocrinology, Baylor College of Medicine Department of Pediatrics, Houston, Texas, USA
| | - William E Winter
- Department of Pathology, University of Florida, Gainesville, Florida, USA
| | | | - Raymond Kreienkamp
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Maria J Redondo
- Texas Children's Hospital—Pediatric Diabetes & Endocrinology, Baylor College of Medicine Department of Pediatrics, Houston, Texas, USA
| | - Mustafa Tosur
- Texas Children's Hospital—Pediatric Diabetes & Endocrinology, Baylor College of Medicine Department of Pediatrics, Houston, Texas, USA
- Children’s Nutrition Research Center, USDA-ARS, Houston, Texas, USA
| |
Collapse
|
2
|
Dye TDV, Quiñones Tavárez Z, Rivera I, Cardona Cordero N. Social determinants of participation in genetic research among Puerto Ricans and in the Puerto Rican diaspora. Soc Sci Med 2024; 362:117437. [PMID: 39461167 DOI: 10.1016/j.socscimed.2024.117437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
Puerto Ricans are underrepresented in genetic research. This underrepresentation denies Puerto Ricans the benefit from therapeutic developments that could mitigate health disparities arising from conditions for which genetically-derived treatments exist. The Puerto Rican diaspora, especially post-2017 due to economic and environmental crises, has expanded within the USA. Prior research suggests that Latin American diaspora communities are less likely to participate in genetic research. We hypothesized, specifically, that the Puerto Rican diaspora in the USA would be less likely to participate in genetic research than would Puerto Ricans in their homeland's archipelago, and that accounting for social and cultural determinants related to the diaspora experience would mitigate this disparity. We implemented an analytical cross-sectional study of archipelago-residing Puerto Ricans and of the USA-residing diaspora to evaluate this hypothesis. With 1582 Puerto Ricans (723 in Puerto Rico, 859 in the USA), we found that while most participants would participate in genetic research, participation rates varied significantly by diaspora status. Puerto Ricans born and living in the USA were initially more likely to decline participation compared to those in Puerto Rico (OR = 1.54, p < 0.01). However, once adjusted for social and cultural variables, this difference was eliminated (aOR = 1.08, p = n.s.). The factors influencing non-participation include oppression, discrimination, distrust, and social determinants, aligning with the theory of minoritization. An important community in the USA and in the world, Puerto Ricans have the right to participate in well-conducted research and to benefit from its findings, particularly around topics that could help address existing disparities in health outcomes.
Collapse
Affiliation(s)
- Timothy De Ver Dye
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| | - Zahira Quiñones Tavárez
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| | - Ivelisse Rivera
- Department of Obstetrics and Gynecology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA.
| | | |
Collapse
|
3
|
Sanyal D. Exploring the genetic basis of childhood monogenic diabetes. World J Diabetes 2024; 15:1829-1832. [PMID: 39280182 PMCID: PMC11372639 DOI: 10.4239/wjd.v15.i9.1829] [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: 12/18/2023] [Revised: 05/29/2024] [Accepted: 06/28/2024] [Indexed: 08/27/2024] Open
Abstract
Monogenic diabetes is caused by one or even more genetic variations, which may be uncommon yet have a significant influence and cause diabetes at an early age. Monogenic diabetes affects 1% to 5% of children, and early detection and genetically focused treatment of neonatal diabetes and maturity-onset diabetes of the young can significantly improve long-term health and well-being. The etiology of monogenic diabetes in childhood is primarily attributed to genetic variations affecting the regulatory genes responsible for beta-cell activity. In rare instances, mutations leading to severe insulin resistance can also result in the development of diabetes. Individuals diagnosed with specific types of monogenic diabetes, which are commonly found, can transition from insulin therapy to sulfonylureas, provided they maintain consistent regulation of their blood glucose levels. Scientists have successfully devised materials and methodologies to distinguish individuals with type 1 or 2 diabetes from those more prone to monogenic diabetes. Genetic screening with appropriate findings and interpretations is essential to establish a prognosis and to guide the choice of therapies and management of these interrelated ailments. This review aims to design a comprehensive literature summarizing genetic insights into monogenetic diabetes in children and adolescents as well as summarizing their diagnosis and management.
Collapse
Affiliation(s)
- Debmalya Sanyal
- Department of Endocrinology, KPC Medical College, Kolkata Pin 700032, West Bengal, India
- Department of Endocrinology, NH RTIICS, Kolkata Pin 700099, West Bengal, India
- School of Medicine, University of New Castle, Callaghan NSW 2308, Australia
| |
Collapse
|
4
|
Samhani C, Guerci B, Larose C. Discovery of a TRMT10A mutation in a case of atypical diabetes: Case report. DIABETES & METABOLISM 2024; 50:101572. [PMID: 39243962 DOI: 10.1016/j.diabet.2024.101572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024]
Abstract
It is notable that monogenic forms of diabetes are exceedingly uncommon, with only 28 genes thus far identified. Such conditions frequently result in the dysfunction of pancreatic cells responsible for insulin production. Mutation in the TRMT10A gene leads to a rare genetic disease that is associated with endocrine and metabolic disorders, including diabetes and short stature. This article presents a review of the existing literature on the subject, describing the association between TRMT10A gene mutation and diabetes. It also presents the clinical case of a young girl with type 1 diabetes and facial dysmorphia. TRMT10A gene mutation has been linked to syndromic juvenile diabetes in a manner analogous to Wolfram's syndrome. This form of diabetes, which manifests in early childhood and is associated with microcephaly, epilepsy and intellectual disability, is caused by mutations in the gene for homolog A of tRNA methyltransferase 10 (TRMT10A). This emphasizes the importance of using a targeted panel to recognize previously unidentified monogenic diabetes among early-onset non-insulin-dependent diabetes in the absence of obesity and autoimmunity. In view of the aforementioned data, it is recommended that TRMT10A sequencing be considered in children or adults with early-onset diabetes and a history of intellectual disability, microcephaly and epilepsy.
Collapse
Affiliation(s)
- C Samhani
- Department of Endocrinology, Diabetology, and Nutrition, Brabois Adult Hospital, University of Lorraine, Vandoeuvre-lès-Nancy, France
| | - B Guerci
- Department of Endocrinology, Diabetology, and Nutrition, Brabois Adult Hospital, University of Lorraine, Vandoeuvre-lès-Nancy, France; Faculty of Medicine, University of Lorraine, Vandoeuvre-lès-Nancy, France
| | - C Larose
- Faculty of Medicine, University of Lorraine, Vandoeuvre-lès-Nancy, France; Department of Urology, University Hospital, Nancy, France.
| |
Collapse
|
5
|
Welsch S, Harvengt A, Gallo P, Martin M, Beckers D, Mouraux T, Seret N, Lebrethon MC, Helaers R, Brouillard P, Vikkula M, Lysy PA. A New Tool to Identify Pediatric Patients with Atypical Diabetes Associated with Gene Polymorphisms. Diabetes Metab J 2024; 48:949-959. [PMID: 38523249 PMCID: PMC11449816 DOI: 10.4093/dmj.2023.0166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/25/2023] [Indexed: 03/26/2024] Open
Abstract
BACKGRUOUND Recent diabetes subclassifications have improved the differentiation between patients with type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus despite several overlapping features, yet without considering genetic forms of diabetes. We sought to facilitate the identification of monogenic diabetes by creating a new tool that we validated in a pediatric maturity-onset diabetes of the young (MODY) cohort. METHODS We first created the DIAgnose MOnogenic DIAbetes (DIAMODIA) criteria based on the pre-existing, but incomplete, MODY calculator. This new score is composed of four strong and five weak criteria, with patients having to display at least one weak and one strong criterion. RESULTS The effectiveness of the DIAMODIA criteria was evaluated in two patient cohorts, the first consisting of patients with confirmed MODY diabetes (n=34) and the second of patients with T1DM (n=390). These DIAMODIA criteria successfully detected 100% of MODY patients. Multiple correspondence analysis performed on the MODY and T1DM cohorts enabled us to differentiate MODY patients from T1DM. The three most relevant variables to distinguish a MODY from T1DM profile were: lower insulin-dose adjusted A1c score ≤9, glycemic target-adjusted A1c score ≤4.5, and absence of three anti-islet cell autoantibodies. CONCLUSION We validated the DIAMODIA criteria, as it effectively identified all monogenic diabetes patients (MODY cohort) and succeeded to differentiate T1DM from MODY patients. The creation of this new and effective tool is likely to facilitate the characterization and therapeutic management of patients with atypical diabetes, and promptly referring them for genetic testing which would markedly improve clinical care and counseling, as well.
Collapse
Affiliation(s)
- Sophie Welsch
- Pediatrics Unit, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Antoine Harvengt
- Pediatrics Unit, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Paola Gallo
- Pediatric Endocrinology Unit, Saint-Luc University Clinics, Brussels, Belgium
| | - Manon Martin
- Louvain Institute of Biomolecular Science and Technology (IBST) Unit, UCLouvain, Brussels, Belgium
| | - Dominique Beckers
- Pediatric Endocrinology and Diabetology Unit, CHU-UCL Namur sites Saint-Elisabeth and Mont-Godinne, Namur, Belgium
| | - Thierry Mouraux
- Pediatric Endocrinology and Diabetology Unit, CHU-UCL Namur sites Saint-Elisabeth and Mont-Godinne, Namur, Belgium
| | - Nicole Seret
- Pediatric Endocrinology and Diabetology Unit, Clinique CHC MontLégia (CHC MontLégia Clinic), Liège, Belgium
| | | | - Raphaël Helaers
- Human Molecular Genetics, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Pascal Brouillard
- Human Molecular Genetics, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Philippe A. Lysy
- Pediatrics Unit, Institute for Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Pediatric Endocrinology Unit, Saint-Luc University Clinics, Brussels, Belgium
| |
Collapse
|
6
|
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
|
7
|
Vourdoumpa A, Paltoglou G, Mertzanian A, Sertedaki A, Sakou II, Karanasios S, Karavanaki K, Charmandari E. Challenges in the management of patients with HNF1B MODY and multisystem manifestations: the cases of two adolescent boys. Hormones (Athens) 2024; 23:439-445. [PMID: 38980656 DOI: 10.1007/s42000-024-00580-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 06/25/2024] [Indexed: 07/10/2024]
Abstract
INTRODUCTION Hepatocyte nuclear factor-1 beta (HNF1B) encodes a homeodomain-containing transcription factor, which is expressed early in embryogenesis and is involved in the development of multiple tissues and organs. HNF1B mutations cause complex multisystem disorders, with renal developmental disease and maturity onset diabetes of the young (HNF1B MODY), a rare cause of diabetes mellitus, being representative features. METHODS We present two adolescent boys from different socioeconomic backgrounds who were diagnosed with genetically confirmed HNF1B MODY following hospitalization for diabetic ketoacidosis in the first case and after diagnostic work-up due to impaired glucose tolerance in the second case. Multisystem manifestations, including pancreatic hypoplasia and early-onset diabetes mellitus (DM), renal cysts, hypomagnesemia, hyperuricemia, liver and biliary impairment, genital tract malformations, and primary hyperparathyroidism were also present, strongly suggesting HNF1B MODY. RESULTS The first patient was treated with subcutaneous insulin but was lost to follow-up due to social reasons. Conversely, early diagnosis in the second patient allowed the management of multisystem defects by a multidisciplinary team of experts. Moreover, manifestation of HNF1B MODY in the form of diabetic ketoacidosis was prevented and a structured diabetes training program has proven successful in regulating glycemic control, postponing the necessity for insulin treatment. CONCLUSION Early genetic work-up of patients with dysglycemia associated with a specific phenotype suggestive of HNF1B MODY is extremely important in the care of children and adolescents with diabetes since it ensures that early and optimal management is initiated, thereby preventing the onset of life-threatening diabetic ketoacidosis and other multisystem complications and/or comorbidities.
Collapse
Affiliation(s)
- Aikaterini Vourdoumpa
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece
- Diabetes and Metabolism Clinic, Second Department of Pediatrics, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, 11527, Greece
| | - George Paltoglou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece.
- Diabetes and Metabolism Clinic, Second Department of Pediatrics, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, 11527, Greece.
| | - Anny Mertzanian
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece
| | - Amalia Sertedaki
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece
| | - Irini-Ikbale Sakou
- Diabetes and Metabolism Clinic, Second Department of Pediatrics, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, 11527, Greece
| | - Spyridon Karanasios
- Diabetes and Metabolism Clinic, Second Department of Pediatrics, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, 11527, Greece
| | - Kyriaki Karavanaki
- Diabetes and Metabolism Clinic, Second Department of Pediatrics, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, 11527, Greece
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
| |
Collapse
|
8
|
Wang X, Cheng W, Wang Z, Liu C, Deng A, Li J. Chinese carrier of the HNF1A p.Gln444fs variant exhibits enhanced response to sulfonylureas. Heliyon 2024; 10:e35112. [PMID: 39170165 PMCID: PMC11336406 DOI: 10.1016/j.heliyon.2024.e35112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Background We assessed the response to sulfonylureas and the functional characteristics of HNF1A mutations in patients with maturity-onset diabetes of the young type 3 (MODY3). Methods We recruited a family with suspected MODY in this study, and gene sequencing (whole-exome sequencing) was used to screen germline mutations. Luciferase reporter assays were used to evaluate the activity of the mutated genes. Results Heterozygous HNF1A variant (NM_000545.8:c.1330_1331del, p.Gln444fs) was identified in the proband and was not found in his father, grandmother, and nonrelated healthy controls. The mutant protein had 552 amino acids, 110 fewer than the wild type protein. Furthermore, the amino acid sequence was completely different between the mutant protein and the wild type protein starting from the 444th amino acid. Luciferase reporter assays revealed that the variant had impaired HNF4A promoter-regulation activity. The patient did not achieve good hypoglycemic effects during long-term treatment with insulin and metformin. The effect of hypoglycemic treatment was highly significant after the addition of sulfonylurea drugs. Conclusions The HNF1A p.Gln444fs variant associated with MODY3, and most likely a truncated protein, impaired HNF1A transcriptional activity. The variant carrier experienced an enhanced response to sulfonylureas.
Collapse
Affiliation(s)
- Xiufang Wang
- Department of Pain, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenzhuo Cheng
- Department of Pediatric, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhongjing Wang
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chao Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Aiping Deng
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Juyi Li
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
9
|
Baccetti F, Crisafulli C, Andreozzi F, Mannino GC, Nicolucci A, Michelli A, Miranda C, Candido R, Di Bartolo P, Di Cianni G, Russo GT, Mannino D. Profiles of sulfonylurea use in Diabetes Mellitus type 2: an analysis of clinical practice over the last 10 years. Diabetes Res Clin Pract 2024; 214:111781. [PMID: 39002933 DOI: 10.1016/j.diabres.2024.111781] [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: 03/07/2024] [Revised: 06/17/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
AIMS Describing the evolution over time in the use of sulfonylureas (SUs) and the characteristics of patients at first prescription and at interruption of treatment with SUs. METHODS Retrospective evaluation of data from the Italian Association of Diabetologists (AMD) Annals registry (2010-2020), about T2D patients who started treatment with SUs. The longitudinal probability of remaining on SUs was estimated by Kaplan Meier survival curves. RESULTS SU prescription decreased from 30.7 % (2010) to 12.9 % (2020). Patients started on SU were 68.2 ± 11.2 years old, mostly males (55.5 %), with diabetes duration = 10.1 ± 8.3 years, BMI = 29.7 ± 5.5 kg/m2, and HbA1c = 8.3 ± 1.7 % [67 mmol/mol]. After one year, the probability of staying on SU was 85.4 %, 75.9 % after two years, 68.2 % after 3 years, 56.6 % after 5 years. Patients who discontinued SUs had higher BMI and HbA1c, were younger, more often males and treated with insulin. Over time, the percentage of subjects switched to metformin, DPP4i, SGLT2i, and GLP1RA increased, whereas use of glinides, glitazones, acarbose and insulin declined. CONCLUSIONS These data suggest a consensus, slowly, but increasingly aligning with the current National indications of dismissing SUs for the treatment of T2D. The new drugs for diabetes should represent a preferable choice in all patients who do not have specific contraindications.
Collapse
Affiliation(s)
| | | | - Francesco Andreozzi
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy; Research Centre for the Prevention and Treatment of Metabolic Diseases (CR METDIS), University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Gaia Chiara Mannino
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy.
| | - Antonio Nicolucci
- CORESEARCH-Center for Outcomes Research and Clinical Epidemiology, Pescara, Italy
| | - Andrea Michelli
- Department of Internal Medicine, SC Diabetes and Center for Treatment of Diabetic Foot, Monfalcone, Gorizia, Italy
| | - Cesare Miranda
- Clinic of Endocrinology and Metabolic Diseases, Pordenone Hospital, Pordenone, Italy
| | - Riccardo Candido
- Diabetes Center, ASUGI University Hospital Giuliano Isontina, Trieste, Italy
| | - Paolo Di Bartolo
- Diabetes Unit, Local Healthcare Authority of Romagna, Ravenna, Italy
| | - Graziano Di Cianni
- ASL North-West Tuscany, Diabetes and Metabolic Diseases, Livorno Hospital, Livorno, Italy
| | - Giuseppina Tiziana Russo
- Department of Clinical and Experimental Medicine, University Hospital G. Martino, Messina, Italy
| | - Domenico Mannino
- Section of Endocrinology and Diabetes, Bianchi Melacrino Morelli Hospital, Reggio Calabria, Italy; AMD Annals Initiative, AMD Foundation, Rome, Italy
| |
Collapse
|
10
|
Naylor RN, Patel KA, Kettunen JLT, Männistö JME, Støy J, Beltrand J, Polak M, Vilsbøll T, Greeley SAW, Hattersley AT, Tuomi T. Precision treatment of beta-cell monogenic diabetes: a systematic review. COMMUNICATIONS MEDICINE 2024; 4:145. [PMID: 39025920 PMCID: PMC11258280 DOI: 10.1038/s43856-024-00556-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Beta-cell monogenic forms of diabetes have strong support for precision medicine. We systematically analyzed evidence for precision treatments for GCK-related hyperglycemia, HNF1A-, HNF4A- and HNF1B-diabetes, and mitochondrial diabetes (MD) due to m.3243 A > G variant, 6q24-transient neonatal diabetes mellitus (TND) and SLC19A2-diabetes. METHODS The search of PubMed, MEDLINE, and Embase for individual and group level data for glycemic outcomes using inclusion (English, original articles written after 1992) and exclusion (VUS, multiple diabetes types, absent/aggregated treatment effect measures) criteria. The risk of bias was assessed using NHLBI study-quality assessment tools. Data extracted from Covidence were summarized and presented as descriptive statistics in tables and text. RESULTS There are 146 studies included, with only six being experimental studies. For GCK-related hyperglycemia, the six studies (35 individuals) assessing therapy discontinuation show no HbA1c deterioration. A randomized trial (18 individuals per group) shows that sulfonylureas (SU) were more effective in HNF1A-diabetes than in type 2 diabetes. Cohort and case studies support SU's effectiveness in lowering HbA1c. Two cross-over trials (each with 15-16 individuals) suggest glinides and GLP-1 receptor agonists might be used in place of SU. Evidence for HNF4A-diabetes is limited. Most reported patients with HNF1B-diabetes (N = 293) and MD (N = 233) are on insulin without treatment studies. Limited data support oral agents after relapse in 6q24-TND and for thiamine improving glycemic control and reducing/eliminating insulin requirement in SLC19A2-diabetes. CONCLUSION There is limited evidence, and with moderate or serious risk of bias, to guide monogenic diabetes treatment. Further evidence is needed to examine the optimum treatment in monogenic subtypes.
Collapse
Affiliation(s)
- Rochelle N Naylor
- Departments of Pediatrics and Medicine, University of Chicago, Chicago, IL, USA
| | - Kashyap A Patel
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Exeter, Devon, UK
| | - Jarno L T Kettunen
- Helsinki University Hospital, Abdominal Centre/Endocrinology, Helsinki, Finland
- Folkhalsan Research Center, Helsinki, Finland
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Jonna M E Männistö
- Departments of Pediatrics and Clinical Genetics, Kuopio University Hospital, Kuopio, Finland
- Department of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Julie Støy
- Steno diabetes center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Jacques Beltrand
- APHP Centre Hôpital Necker Enfants Malades Université Paris Cité, Paris, France
| | - Michel Polak
- Inserm U1016 Institut Cochin, Paris, France
- Department of Pediatric Endocrinology, Gynecology and Diabetology, Hôpital Universitaire Necker Enfants Malades, Paris, France
- Université Paris Cité, Paris, France
| | - Tina Vilsbøll
- Department of Clinical Medicine, University of Copenhagen, København, Denmark
| | - Siri A W Greeley
- Departments of Pediatrics and Medicine, University of Chicago, Chicago, IL, USA
| | - Andrew T Hattersley
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Exeter, Devon, UK
| | - Tiinamaija Tuomi
- Helsinki University Hospital, Abdominal Centre/Endocrinology, Helsinki, Finland.
- Folkhalsan Research Center, Helsinki, Finland.
- Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland.
- Lund University Diabetes Center, Malmo, Sweden.
| |
Collapse
|
11
|
Zečević K, Volčanšek Š, Katsiki N, Rizzo M, Milardović TM, Stoian AP, Banach M, Muzurović E. Maturity-onset diabetes of the young (MODY) - in search of ideal diagnostic criteria and precise treatment. Prog Cardiovasc Dis 2024; 85:14-25. [PMID: 38513726 DOI: 10.1016/j.pcad.2024.03.004] [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: 03/17/2024] [Accepted: 03/17/2024] [Indexed: 03/23/2024]
Abstract
Maturity-onset diabetes of the young (MODY) is a spectrum of clinically heterogenous forms of monogenic diabetes mellitus characterized by autosomal dominant inheritance, onset at a young age, and absence of pancreatic islets autoimmunity. This rare form of hyperglycemia, with clinical features overlapping with type 1 and type 2 diabetes mellitus, has 14 subtypes with differences in prevalence and complications occurrence which tailor therapeutic approach. MODY phenotypes differ based on the gene involved, gene penetrance and expressivity. While MODY 2 rarely leads to diabetic complications and is easily managed with lifestyle interventions alone, more severe subtypes, such as MODY 1, 3, and 6, require an individualized treatment approach to maintain a patient's quality of life and prevention of complications. This review summarizes current evidence on the presentation, diagnosis, and management of MODY, an example of a genetic cause of hyperglycemia that calls for a precision medicine approach.
Collapse
Affiliation(s)
- Ksenija Zečević
- Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Špela Volčanšek
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia; Medical Faculty Ljubljana, Ljubljana, Slovenia
| | - Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece; School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy; Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Tanja Miličević Milardović
- Internal Medicine Department, Endocrinology, Diabetology, and Metabolism Division, University Hospital of Split, Split, Croatia; University of Split School of Medicine, Split, Croatia
| | - Anca Pantea Stoian
- Diabetes, Nutrition and Metabolic diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Łódź, Lodz, Poland; Department of Cardiology and Adult Congenital Heart Diseases, Polish Mother's Memorial Hospital Research Institute, Łódź, Poland; Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emir Muzurović
- Faculty of Medicine, University of Montenegro, Podgorica, Montenegro; Department of Internal Medicine, Endocrinology Section, Clinical Center of Montenegro, Podgorica, Montenegro.
| |
Collapse
|
12
|
Li H, Li W, Li D, Yuan L, Xu Y, Su P, Wu L, Zhang Z. Based on systematic druggable genome-wide Mendelian randomization identifies therapeutic targets for diabetes. Front Endocrinol (Lausanne) 2024; 15:1366290. [PMID: 38915894 PMCID: PMC11194396 DOI: 10.3389/fendo.2024.1366290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 05/28/2024] [Indexed: 06/26/2024] Open
Abstract
Purpose Diabetes and its complications cause a heavy burden of disease worldwide. In recent years, Mendelian randomization (MR) has been widely used to discover the pathogenesis and epidemiology of diseases, as well as to discover new therapeutic targets. Therefore, based on systematic "druggable" genomics, we aim to identify new therapeutic targets for diabetes and analyze its pathophysiological mechanisms to promote its new therapeutic strategies. Material and method We used double sample MR to integrate the identified druggable genomics to evaluate the causal effect of quantitative trait loci (eQTLs) expressed by druggable genes in blood on type 1 and 2 diabetes (T1DM and T2DM). Repeat the study using different data sources on diabetes and its complications to verify the identified genes. Not only that, we also use Bayesian co-localization analysis to evaluate the posterior probabilities of different causal variations, shared causal variations, and co-localization probabilities to examine the possibility of genetic confounding. Finally, using diabetes markers with available genome-wide association studies data, we evaluated the causal relationship between established diabetes markers to explore possible mechanisms. Result Overall, a total of 4,477 unique druggable genes have been gathered. After filtering using methods such as Bonferroni significance (P<1.90e-05), the MR Steiger directionality test, Bayesian co-localization analysis, and validation with different datasets, Finally, 7 potential druggable genes that may affect the results of T1DM and 7 potential druggable genes that may affect the results of T2DM were identified. Reverse MR suggests that C4B may play a bidirectional role in the pathogenesis of T1DM, and none of the other 13 target genes have a reverse causal relationship. And the 7 target genes in T2DM may each affect the biomarkers of T2DM to mediate the pathogenesis of T2DM. Conclusion This study provides genetic evidence supporting the potential therapeutic benefits of targeting seven druggable genes, namely MAP3K13, KCNJ11, REG4, KIF11, CCNE2, PEAK1, and NRBP1, for T2DM treatment. Similarly, targeting seven druggable genes, namely ERBB3, C4B, CD69, PTPN22, IL27, ATP2A1, and LT-β, has The potential therapeutic benefits of T1DM treatment. This will provide new ideas for the treatment of diabetes and also help to determine the priority of drug development for diabetes.
Collapse
Affiliation(s)
- Hu Li
- Emergency Department, Binzhou Medical University Hospital, Binzhou, China
| | - Wei Li
- Urology Department, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Dongyang Li
- Internal Medicine-Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Lijuan Yuan
- Emergency Department, Binzhou Medical University Hospital, Binzhou, China
| | - Yucheng Xu
- Department of Critical Care Medicine, Jinan Central Hospital, Jinan, China
| | - Pengtao Su
- Emergency Department, Binzhou Medical University Hospital, Binzhou, China
| | - Liqiang Wu
- Emergency Department, Binzhou Medical University Hospital, Binzhou, China
| | - Zhiqiang Zhang
- Emergency Department, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
13
|
Fan Y, Chow E, Lim CKP, Hou Y, Tsoi STF, Fan B, Lau ESH, Kong APS, Ma RCW, Wu H, Chan JCN, Luk AOY. Comparison of β-Cell Function and Insulin Sensitivity Between Normal-Weight and Obese Chinese With Young-Onset Type 2 Diabetes. Diabetes 2024; 73:953-963. [PMID: 38506952 DOI: 10.2337/db23-0966] [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: 12/07/2023] [Accepted: 03/17/2024] [Indexed: 03/22/2024]
Abstract
Normal-weight individuals with usual-onset type 2 diabetes have reduced β-cell function and greater insulin sensitivity compared with their obese counterparts. The relative contribution of β-cell dysfunction and insulin resistance to young-onset type 2 diabetes (YOD) among normal-weight individuals is not well established. In 44 individuals with YOD (24 with normal weight and 20 with obesity) and 24 healthy control individuals with normoglycemia (12 with normal weight and 12 with obesity), we conducted 2-h 12 mmol/L hyperglycemic clamps to measure acute (0-10 min) and steady-state (100-120 min) insulin and C-peptide responses, as well as insulin sensitivity index. Normal-weight individuals with YOD had lower acute insulin response, steady-state insulin and C-peptide responses, and a higher insulin sensitivity index compared with their obese counterparts with YOD. Compared with BMI-matched healthy control individuals, normal-weight individuals with YOD had lower acute and steady-state insulin and C-peptide responses but a similar insulin sensitivity index. The impairment of steady-state β-cell response relative to healthy control individuals was more pronounced in normal-weight versus obese individuals with YOD. In conclusion, normal-weight Chinese with YOD exhibited worse β-cell function but preserved insulin sensitivity relative to obese individuals with YOD and BMI-matched healthy individuals with normoglycemia. The selection of glucose-lowering therapy should account for pathophysiological differences underlying YOD between normal-weight and obese individuals. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Yingnan Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Cadmon K P Lim
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Yong Hou
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Sandra T F Tsoi
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Baoqi Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Eric S H Lau
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Alice P S Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Hongjiang Wu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Andrea O Y Luk
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| |
Collapse
|
14
|
Al Sadi K, Balachandran W. Leveraging a 7-Layer Long Short-Term Memory Model for Early Detection and Prevention of Diabetes in Oman: An Innovative Approach. Bioengineering (Basel) 2024; 11:379. [PMID: 38671800 PMCID: PMC11048439 DOI: 10.3390/bioengineering11040379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/28/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
This study develops a 7-layer Long Short-Term Memory (LSTM) model to enhance early diabetes detection in Oman, aligning with the theme of 'Artificial Intelligence in Healthcare'. The model focuses on addressing the increasing prevalence of Type 2 diabetes, projected to impact 23.8% of Oman's population by 2050. It employs LSTM neural networks to manage factors contributing to this rise, including obesity and genetic predispositions, and aims to bridge the gap in public health awareness and prevention. The model's performance is evaluated through various metrics. It achieves an accuracy of 99.40%, specificity and sensitivity of 100% for positive cases, a recall of 99.34% for negative cases, an F1 score of 96.24%, and an AUC score of 94.51%. These metrics indicate the model's capability in diabetes detection. The implementation of this LSTM model in Oman's healthcare system is proposed to enhance early detection and prevention of diabetes. This approach reflects an application of AI in addressing a significant health concern, with potential implications for similar healthcare challenges relating to globally diagnostic capabilities, representing a significant leap forward in healthcare technology in Oman.
Collapse
Affiliation(s)
- Khoula Al Sadi
- Department of Electronic and Electrical Engineering Research, Brunel University London, Uxbridge UB8 3PH, UK;
- Information Technology Department, University of Technology and Applied Sciences-Al-Mussanha, P.O. Box 13, Muladdah 314, Oman
| | - Wamadeva Balachandran
- Department of Electronic and Electrical Engineering Research, Brunel University London, Uxbridge UB8 3PH, UK;
| |
Collapse
|
15
|
Chan JC, O CK, Luk AO. Young-Onset Diabetes in East Asians: From Epidemiology to Precision Medicine. Endocrinol Metab (Seoul) 2024; 39:239-254. [PMID: 38626908 PMCID: PMC11066447 DOI: 10.3803/enm.2024.1968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 05/03/2024] Open
Abstract
Precision diagnosis is the keystone of clinical medicine. In East Asians, classical type 1 diabetes is uncommon in patients with youngonset diabetes diagnosed before age of 40, in whom a family history, obesity, and beta-cell and kidney dysfunction are key features. Young-onset diabetes affects one in five Asian adults with diabetes in clinic settings; however, it is often misclassified, resulting in delayed or non-targeted treatment. Complex aetiologies, long disease duration, aggressive clinical course, and a lack of evidence-based guidelines have contributed to variable care standards and premature death in these young patients. The high burden of comorbidities, notably mental illness, highlights the numerous knowledge gaps related to this silent killer. The majority of adult patients with youngonset diabetes are managed as part of a heterogeneous population of patients with various ages of diagnosis. A multidisciplinary care team led by physicians with special interest in young-onset diabetes will help improve the precision of diagnosis and address their physical, mental, and behavioral health. To this end, payors, planners, and providers need to align and re-design the practice environment to gather data systematically during routine practice to elucidate the multicausality of young-onset diabetes, treat to multiple targets, and improve outcomes in these vulnerable individuals.
Collapse
Affiliation(s)
- Juliana C.N. Chan
- Department of Medicine and Therapeutics, Hong Kong Institute of Diabetes and Obesity and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong
| | - Chun-Kwan O
- Department of Medicine and Therapeutics, Hong Kong Institute of Diabetes and Obesity and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong
| | - Andrea O.Y. Luk
- Department of Medicine and Therapeutics, Hong Kong Institute of Diabetes and Obesity and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong
| |
Collapse
|
16
|
Mueller LM, Isaacson A, Wilson H, Salowka A, Tay I, Gong M, Elbarbary NS, Raile K, Spagnoli FM. Heterozygous missense variant in GLI2 impairs human endocrine pancreas development. Nat Commun 2024; 15:2483. [PMID: 38509065 PMCID: PMC10954617 DOI: 10.1038/s41467-024-46740-8] [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: 03/02/2023] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
Missense variants are the most common type of coding genetic variants. Their functional assessment is fundamental for defining any implication in human diseases and may also uncover genes that are essential for human organ development. Here, we apply CRISPR-Cas9 gene editing on human iPSCs to study a heterozygous missense variant in GLI2 identified in two siblings with early-onset and insulin-dependent diabetes of unknown cause. GLI2 is a primary mediator of the Hedgehog pathway, which regulates pancreatic β-cell development in mice. However, neither mutations in GLI2 nor Hedgehog dysregulation have been reported as cause or predisposition to diabetes. We establish and study a set of isogenic iPSC lines harbouring the missense variant for their ability to differentiate into pancreatic β-like cells. Interestingly, iPSCs carrying the missense variant show altered GLI2 transcriptional activity and impaired differentiation of pancreatic progenitors into endocrine cells. RNASeq and network analyses unveil a crosstalk between Hedgehog and WNT pathways, with the dysregulation of non-canonical WNT signaling in pancreatic progenitors carrying the GLI2 missense variant. Collectively, our findings underscore an essential role for GLI2 in human endocrine development and identify a gene variant that may lead to diabetes.
Collapse
Affiliation(s)
- Laura M Mueller
- Centre for Gene Therapy and Regenerative Medicine, King's College London, Great Maze Pond, London, SE1 9RT, United Kingdom
| | - Abigail Isaacson
- Centre for Gene Therapy and Regenerative Medicine, King's College London, Great Maze Pond, London, SE1 9RT, United Kingdom
| | - Heather Wilson
- Centre for Gene Therapy and Regenerative Medicine, King's College London, Great Maze Pond, London, SE1 9RT, United Kingdom
| | - Anna Salowka
- Centre for Gene Therapy and Regenerative Medicine, King's College London, Great Maze Pond, London, SE1 9RT, United Kingdom
| | - Isabel Tay
- Centre for Gene Therapy and Regenerative Medicine, King's College London, Great Maze Pond, London, SE1 9RT, United Kingdom
| | - Maolian Gong
- Department of Pediatric Endocrinology and Diabetology, Charité, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Charité Medical Faculty, Max-Delbrueck-Center for Molecular Medicine (MDC), Berlin, Germany
| | - Nancy Samir Elbarbary
- Department of Pediatrics, Diabetes and Endocrine Unit, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Klemens Raile
- Department of Pediatric Endocrinology and Diabetology, Charité, Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Charité Medical Faculty, Max-Delbrueck-Center for Molecular Medicine (MDC), Berlin, Germany
| | - Francesca M Spagnoli
- Centre for Gene Therapy and Regenerative Medicine, King's College London, Great Maze Pond, London, SE1 9RT, United Kingdom.
| |
Collapse
|
17
|
Zhao J, Chen Y, Ma F, Shu H, Zheng L, Liu Y, Li X, Xu T, Zhou Z, Zhou K. MODY Probability Calculator Is Suitable for MODY Screening in China: A Population-based Study. J Endocr Soc 2024; 8:bvae047. [PMID: 38562131 PMCID: PMC10983078 DOI: 10.1210/jendso/bvae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Indexed: 04/04/2024] Open
Abstract
Context Selecting appropriate individuals for genetic testing is essential due to the optimal treatment for maturity-onset diabetes of the young (MODY). However, how to effectively screen for MODY in China remains unclear. Objective To validate the performance of current screening strategies in selecting patients with MODY based on a nationwide type 2 diabetes cohort. Methods A panel of 14 MODY genes was analyzed from 1911 type 2 diabetes patients who were ages 15 to 35 years. Variants were evaluated according to the American College of Medical Genetics and Genomics guidelines. Based on this cohort, we simulated the 2 most frequently used screening strategies, including the traditional MODY criteria and the MODY probability calculator (MPC), to assess their ability to select patients with MODY. Results From a total of 1911 participants, 42 participants harbored pathogenic/likely pathogenic variants. The performance of the traditional criteria was sensitivity: 19.0%, specificity: 72.9%, positive predictive value (PPV): 1.6%, and missing rate: 81.0%. The optimal cut-off for MPC was 40.7%. Based on this cut-off value, the performance was sensitivity: 54.8%, specificity: 81.0%, PPV: 6.1%, and missing rate: 45.2%. Moreover, hemoglobin A1c, insulin treatment, and family history of diabetes have poor discrimination between MODY and young-onset type 2 diabetes. Conclusion The MPC is better than traditional criteria in terms of both sensitivity and PPV. To ensure more MODY patients benefit from optimal treatment, we therefore suggest that routine genetic testing be performed on all type 2 diabetes patients who are between the ages of 15 and35 years and have MPC probability value over 40.7%.
Collapse
Affiliation(s)
- Jing Zhao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yan Chen
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Fuhui Ma
- Department of Endocrinology and Metabolic Diseases, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Diabetes, Urumqi, 830001, China
| | - Hua Shu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Heping District, Tianjin, 300052, China
| | - Li Zheng
- Key Laboratory of RNA Biology, Center for Big Data Research in Health, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yang Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Tao Xu
- Guangzhou Laboratory, Guangdong Province, Guangdong 510005, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Kaixin Zhou
- Guangzhou Laboratory, Guangdong Province, Guangdong 510005, China
- School of Public Health, Guangzhou Medical University, Xinzao, Panyu District, Guangzhou, 511436, China
| |
Collapse
|
18
|
Guo Y, Chen Y, Liang W, Zeng L, Hu F, Zeng Y, Cong L. A novel mutation in the ABCC8 gene causing maturity-onset diabetes of the young: A case report. Clin Med (Lond) 2024; 24:100033. [PMID: 38513803 PMCID: PMC11091440 DOI: 10.1016/j.clinme.2024.100033] [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: 03/23/2024]
Abstract
A 34-year-old woman was diagnosed with type 1 diabetes mellitus and treated with insulin for 24 years. The patient has a family history of diabetes in three consecutive generations. Her Whole exon sequencing showed a heterozygous mutation in the ABCC8 gene, and it also found some of her relatives to carry this mutation. She was diagnosed with MODY12 and received glimepiride therapy with the achievement of good glycaemic control.
Collapse
Affiliation(s)
- Yifan Guo
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yangli Chen
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Wen Liang
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Lirong Zeng
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Fang Hu
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yingjuan Zeng
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Li Cong
- Department of Endocrinology and Metabolism, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong, China.
| |
Collapse
|
19
|
Bansal V, Winkelmann BR, Dietrich JW, Boehm BO. Whole-exome sequencing in familial type 2 diabetes identifies an atypical missense variant in the RyR2 gene. Front Endocrinol (Lausanne) 2024; 15:1258982. [PMID: 38444585 PMCID: PMC10913019 DOI: 10.3389/fendo.2024.1258982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/10/2024] [Indexed: 03/07/2024] Open
Abstract
Genome-wide association studies have identified several hundred loci associated with type 2 diabetes mellitus (T2DM). Additionally, pathogenic variants in several genes are known to cause monogenic diabetes that overlaps clinically with T2DM. Whole-exome sequencing of related individuals with T2DM is a powerful approach to identify novel high-penetrance disease variants in coding regions of the genome. We performed whole-exome sequencing on four related individuals with T2DM - including one individual diagnosed at the age of 33 years. The individuals were negative for mutations in monogenic diabetes genes, had a strong family history of T2DM, and presented with several characteristics of metabolic syndrome. A missense variant (p.N2291D) in the type 2 ryanodine receptor (RyR2) gene was one of eight rare coding variants shared by all individuals. The variant was absent in large population databases and affects a highly conserved amino acid located in a mutational hotspot for pathogenic variants in Catecholaminergic polymorphic ventricular tachycardia (CPVT). Electrocardiogram data did not reveal any cardiac abnormalities except a lower-than-normal resting heart rate (< 60 bpm) in two individuals - a phenotype observed in CPVT individuals with RyR2 mutations. RyR2-mediated Ca2+ release contributes to glucose-mediated insulin secretion and pathogenic RyR2 mutations cause glucose intolerance in humans and mice. Analysis of glucose tolerance testing data revealed that missense mutations in a CPVT mutation hotspot region - overlapping the p.N2291D variant - are associated with complete penetrance for glucose intolerance. In conclusion, we have identified an atypical missense variant in the RyR2 gene that co-segregates with diabetes in the absence of overt CPVT.
Collapse
Affiliation(s)
- Vikas Bansal
- Department of Pediatrics, University of California San Diego, La Jolla, CA, United States
- Institute of Genomic Medicine, University of California San Diego, La Jolla, CA, United States
| | | | - Johannes W Dietrich
- Diabetes, Endocrinology and Metabolism Section, Department of Internal Medicine I, St. Josef Hospital, Ruhr University Hospitals, Bochum, Germany
- Diabetes Center Bochum-Hattingen, St. Elisabeth-Hospital Blankenstein, Hattingen, Germany
- Center for Rare Endocrine Diseases, Ruhr Center for Rare Diseases (CeSER), Ruhr University Bochum and Witten/Herdecke University, Bochum, Germany
- Center for Diabetes Technology, Catholic Hospitals Bochum, Bochum, Germany
| | - Bernhard O Boehm
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
20
|
Kwak SH, Srinivasan S, Chen L, Todd J, Mercader JM, Jensen ET, Divers J, Mottl AK, Pihoker C, Gandica RG, Laffel LM, Isganaitis E, Haymond MW, Levitsky LL, Pollin TI, Florez JC, Flannick J. Genetic architecture and biology of youth-onset type 2 diabetes. Nat Metab 2024; 6:226-237. [PMID: 38278947 PMCID: PMC10896722 DOI: 10.1038/s42255-023-00970-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 12/20/2023] [Indexed: 01/28/2024]
Abstract
The prevalence of youth-onset type 2 diabetes (T2D) and childhood obesity has been rising steadily1, producing a growing public health concern1 that disproportionately affects minority groups2. The genetic basis of youth-onset T2D and its relationship to other forms of diabetes are unclear3. Here we report a detailed genetic characterization of youth-onset T2D by analysing exome sequences and common variant associations for 3,005 individuals with youth-onset T2D and 9,777 adult control participants matched for ancestry, including both males and females. We identify monogenic diabetes variants in 2.4% of individuals and three exome-wide significant (P < 2.6 × 10-6) gene-level associations (HNF1A, MC4R, ATXN2L). Furthermore, we report rare variant association enrichments within 25 gene sets related to obesity, monogenic diabetes and β-cell function. Many youth-onset T2D associations are shared with adult-onset T2D, but genetic risk factors of all frequencies-and rare variants in particular-are enriched within youth-onset T2D cases (5.0-fold increase in the rare variant and 3.4-fold increase in common variant genetic liability relative to adult-onset cases). The clinical presentation of participants with youth-onset T2D is influenced in part by the frequency of genetic risk factors within each individual. These findings portray youth-onset T2D as a heterogeneous disease situated on a spectrum between monogenic diabetes and adult-onset T2D.
Collapse
Affiliation(s)
- Soo Heon Kwak
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shylaja Srinivasan
- Division of Pediatric Endocrinology, University of California at San Francisco, San Francisco, CA, USA
| | - Ling Chen
- Center for Genomic Medicine, and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jennifer Todd
- Division of Pediatric Endocrinology, Department of Pediatrics, University of Vermont, Burlington, VT, USA
| | - Josep M Mercader
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Elizabeth T Jensen
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jasmin Divers
- Department of Foundations of Medicine, NYU Langone Health, New York, NY, USA
| | - Amy K Mottl
- Division of Nephrology and Hypertension, University of North Carolina, Chapel Hill, NC, USA
| | - Catherine Pihoker
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Rachelle G Gandica
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Lori M Laffel
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | | | - Morey W Haymond
- Department of Pediatrics, Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Lynne L Levitsky
- Pediatric Endocrine Division, Department of Pediatrics, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, USA
| | - Toni I Pollin
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jose C Florez
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, and Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jason Flannick
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
21
|
Morgan NG. Insulitis in human type 1 diabetes: lessons from an enigmatic lesion. Eur J Endocrinol 2024; 190:lvae002. [PMID: 38231086 PMCID: PMC10824273 DOI: 10.1093/ejendo/lvae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/14/2023] [Accepted: 12/18/2023] [Indexed: 01/18/2024]
Abstract
Type 1 diabetes is caused by a deficiency of insulin secretion which has been considered traditionally as the outcome of a precipitous decline in the viability of β-cells in the islets of Langerhans, brought about by autoimmune-mediated attack. Consistent with this, various classes of lymphocyte, as well as cells of the innate immune system have been found in association with islets during disease progression. However, analysis of human pancreas from subjects with type 1 diabetes has revealed that insulitis is often less intense than in equivalent animal models of the disease and can affect many fewer islets than expected, at disease onset. This is especially true in subjects developing type 1 diabetes in, or beyond, their teenage years. Such studies imply that both the phenotype and the number of immune cells present within insulitic lesions can vary among individuals in an age-dependent manner. Additionally, the influent lymphocytes are often mainly arrayed peripherally around islets rather than gaining direct access to the endocrine cell core. Thus, insulitis remains an enigmatic phenomenon in human pancreas and this review seeks to explore the current understanding of its likely role in the progression of type 1 diabetes.
Collapse
Affiliation(s)
- Noel G Morgan
- Department of Clinical and Biomedical Science, Islet Biology Exeter (IBEx), Exeter Centre of Excellence in Diabetes (EXCEED), University of Exeter Medical School, Exeter EX2 5DW, United Kingdom
| |
Collapse
|
22
|
Jędrysik M, Wyszomirski K, Różańska-Walędziak A, Grosicka-Maciąg E, Walędziak M, Chełstowska B. The Role of GLP-1, GIP, MCP-1 and IGFBP-7 Biomarkers in the Development of Metabolic Disorders: A Review and Predictive Analysis in the Context of Diabetes and Obesity. Biomedicines 2024; 12:159. [PMID: 38255264 PMCID: PMC10813748 DOI: 10.3390/biomedicines12010159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/29/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Metabolic illnesses, including obesity and type 2 diabetes, have become worldwide epidemics that have an effect on public health. Clinical investigations and further exploration of these mechanisms could lead to innovative, effective, and personalized treatment strategies for individuals. It is important to screen biomarkers in previous studies to discover what is missing. Glucagon-like peptide-1's role in insulin secretion and glucose control highlights its diagnostic and therapeutic potential. Glucose-dependent insulinotropic peptide's influence on postprandial satiety and weight management signifies its importance in understanding metabolic processes. Monocyte chemoattractant protein-1's involvement in inflammation and insulin resistance underlines its value as a diagnostic marker. Insulin-like growth factor-binding protein-7's association with insulin sensitivity and kidney function presents it as a potential target for these diseases' management. In validating these biomarkers, it will be easier to reflect pathophysiological processes, and clinicians will be able to better assess disease severity, monitor disease progression, and tailor treatment strategies. The purpose of the study was to elucidate the significance of identifying novel biomarkers for type 2 diabetes mellitus and obesity, which can revolutionize early detection, risk assessment, and personalized treatment strategies. Standard literature searches of PubMed (MEDLINE), EMBASE, and Cochrane Library were conducted in the year 2023 to identify both original RCTs and recent systematic reviews that have explored the importance of identifying novel biomarkers for T2D and obesity. This search produced 1964 results, and then was reduced to randomized controlled trial and systematic reviews, producing 145 results and 44 results, respectively. Researchers have discovered potential associations between type 2 diabetes mellitus and obesity and the biomarkers glucagon-like peptide-1, glucose-dependent insulinotropic peptide, monocyte chemoattractant protein-1, and insulin-like growth factor-binding protein-7. Understanding the role of those biomarkers in disease pathogenesis offers hope for improving diagnostics, personalized treatment, and prevention strategies.
Collapse
Affiliation(s)
- Malwina Jędrysik
- Department of Biochemistry and Laboratory Diagnostics, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University in Warsaw, 01-938 Warsaw, Poland; (M.J.); (E.G.-M.); (B.C.)
| | - Krzysztof Wyszomirski
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University in Warsaw, 01-938 Warsaw, Poland;
| | - Anna Różańska-Walędziak
- Department of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University in Warsaw, 01-938 Warsaw, Poland;
| | - Emilia Grosicka-Maciąg
- Department of Biochemistry and Laboratory Diagnostics, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University in Warsaw, 01-938 Warsaw, Poland; (M.J.); (E.G.-M.); (B.C.)
| | - Maciej Walędziak
- Department of General, Oncological, Metabolic and Thoracic Surgery, Military Institute of Medicine—National Research Institute, Szaserów 128 St., 04-141 Warsaw, Poland;
| | - Beata Chełstowska
- Department of Biochemistry and Laboratory Diagnostics, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University in Warsaw, 01-938 Warsaw, Poland; (M.J.); (E.G.-M.); (B.C.)
| |
Collapse
|
23
|
Sun HY, Lin XY. Genetic perspectives on childhood monogenic diabetes: Diagnosis, management, and future directions. World J Diabetes 2023; 14:1738-1753. [PMID: 38222792 PMCID: PMC10784795 DOI: 10.4239/wjd.v14.i12.1738] [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: 09/02/2023] [Revised: 10/10/2023] [Accepted: 11/14/2023] [Indexed: 12/14/2023] Open
Abstract
Monogenic diabetes is caused by one or even more genetic variations, which may be uncommon yet have a significant influence and cause diabetes at an early age. Monogenic diabetes affects 1 to 5% of children, and early detection and gene-tically focused treatment of neonatal diabetes and maturity-onset diabetes of the young can significantly improve long-term health and well-being. The etiology of monogenic diabetes in childhood is primarily attributed to genetic variations affecting the regulatory genes responsible for beta-cell activity. In rare instances, mutations leading to severe insulin resistance can also result in the development of diabetes. Individuals diagnosed with specific types of monogenic diabetes, which are commonly found, can transition from insulin therapy to sulfonylureas, provided they maintain consistent regulation of their blood glucose levels. Scientists have successfully devised materials and methodologies to distinguish individuals with type 1 or 2 diabetes from those more prone to monogenic diabetes. Genetic screening with appropriate findings and interpretations is essential to establish a prognosis and to guide the choice of therapies and management of these interrelated ailments. This review aims to design a comprehensive literature summarizing genetic insights into monogenetic diabetes in children and adolescents as well as summarizing their diagnosis and mana-gement.
Collapse
Affiliation(s)
- Hong-Yan Sun
- Department of Endocrine and Metabolic Diseases, Yantaishan Hospital, Yantai 264003, Shandong Province, China
| | - Xiao-Yan Lin
- Department of Endocrine and Metabolic Diseases, Yantaishan Hospital, Yantai 264003, Shandong Province, China
| |
Collapse
|
24
|
Bassyouni M, Mysara M, Wohlers I, Busch H, Saber-Ayad M, El-Hadidi M. A comprehensive analysis of genetic risk for metabolic syndrome in the Egyptian population via allele frequency investigation and Missense3D predictions. Sci Rep 2023; 13:20517. [PMID: 37993469 PMCID: PMC10665412 DOI: 10.1038/s41598-023-46844-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023] Open
Abstract
Diabetes mellitus (DM) represents a major health problem in Egypt and worldwide, with increasing numbers of patients with prediabetes every year. Numerous factors, such as obesity, hyperlipidemia, and hypertension, which have recently become serious concerns, affect the complex pathophysiology of diabetes. These metabolic syndrome diseases are highly linked to genetic variability that drives certain populations, such as Egypt, to be more susceptible to developing DM. Here we conduct a comprehensive analysis to pinpoint the similarities and uniqueness among the Egyptian genome reference and the 1000-genome subpopulations (Europeans, Ad-Mixed Americans, South Asians, East Asians, and Africans), aiming at defining the potential genetic risk of metabolic syndromes. Selected approaches incorporated the analysis of the allele frequency of the different populations' variations, supported by genotypes' principal component analysis. Results show that the Egyptian's reference metabolic genes were clustered together with the Europeans', Ad-Mixed Americans', and South-Asians'. Additionally, 8563 variants were uniquely identified in the Egyptian cohort, from those, two were predicted to cause structural damage, namely, CDKAL1: 6_21065070 (A > T) and PPARG: 3_12351660 (C > T) utilizing the Missense3D database. The former is a protein coding gene associated with Type 2 DM while the latter is a key regulator of adipocyte differentiation and glucose homeostasis. Both variants were detected heterozygous in two different Egyptian individuals from overall 110 sample. This analysis sheds light on the unique genetic traits of the Egyptian population that play a role in the DM high prevalence in Egypt. The proposed analysis pipeline -available through GitHub- could be used to conduct similar analysis for other diseases across populations.
Collapse
Affiliation(s)
- Mahmoud Bassyouni
- Bioinformatics Group, Center for Informatics Sciences (CIS), School of Information Technology and Computer Science (ITCS), Nile University, Giza, Egypt
- Bioscience Research Laboratories Department, MARC for Medical Services and Scientific Research, 6th of October, Jiza, Egypt
| | - Mohamed Mysara
- Bioinformatics Group, Center for Informatics Sciences (CIS), School of Information Technology and Computer Science (ITCS), Nile University, Giza, Egypt
- Microbiology unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Inken Wohlers
- Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology, and Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
- Biomolecular Data Science in Pneumology, Research Center Borstel, 23845, Borstel, Germany
| | - Hauke Busch
- Medical Systems Biology Division, Lübeck Institute of Experimental Dermatology, and Institute for Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
- University Cancer Center Schleswig-Holstein, University Hospital of Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
| | - Maha Saber-Ayad
- Department of Clinical Sciences, College of Medicine, University of Sharjah, 27272, Sharjah, UAE.
- Pharmacology Department, College of Medicine, Cairo University, Cairo, 12613, Egypt.
| | - Mohamed El-Hadidi
- Bioinformatics Group, Center for Informatics Sciences (CIS), School of Information Technology and Computer Science (ITCS), Nile University, Giza, Egypt.
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham Dubai Campus, Dubai, United Arab Emirates.
| |
Collapse
|
25
|
Hocking S, Sumithran P. Individualised prescription of medications for treatment of obesity in adults. Rev Endocr Metab Disord 2023; 24:951-960. [PMID: 37202547 PMCID: PMC10492708 DOI: 10.1007/s11154-023-09808-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/24/2023] [Indexed: 05/20/2023]
Abstract
Obesity continues to increase in prevalence globally, driven by changes in environmental factors which have accelerated the development of obesity in individuals with an underlying predisposition to weight gain. The adverse health effects and increased risk for chronic disease associated with obesity are ameliorated by weight loss, with greater benefits from larger amounts of weight reduction. Obesity is a heterogeneous condition, with the drivers, phenotype and complications differing substantially between individuals. This raises the question of whether treatments for obesity, specifically pharmacotherapy, can be targeted based on individual characteristics. This review examines the rationale and the clinical data evaluating this strategy in adults. Individualised prescribing of obesity medication has been successful in rare cases of monogenic obesity where medications have been developed to target dysfunctions in leptin/melanocortin signalling pathways but has been unsuccessful in polygenic obesity due to a lack of understanding of how the gene variants associated with body mass index affect phenotype. At present, the only factor consistently associated with longer-term efficacy of obesity pharmacotherapy is early weight loss outcome, which cannot inform choice of therapy at the time of medication initiation. The concept of matching a therapy for obesity to the characteristics of the individual is appealing but as yet unproven in randomised clinical trials. With increasing technology allowing deeper phenotyping of individuals, increased sophistication in the analysis of big data and the emergence of new treatments, it is possible that precision medicine for obesity will eventuate. For now, a personalised approach that takes into account the person's context, preferences, comorbidities and contraindications is recommended.
Collapse
Affiliation(s)
- Samantha Hocking
- Faculty of Medicine and Health, University of Sydney, Camperdown, New South Wales, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Priya Sumithran
- Department of Medicine, (St Vincent's Hospital), University of Melbourne, VIC, Fitzroy, Australia.
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia.
- Department of Surgery, Central Clinical School, Monash University, Level 6, 99 Commercial Rd, Melbourne, Victoria, 3004, Australia.
| |
Collapse
|
26
|
Naylor RN, Patel KA, Kettunen JL, Männistö JM, Støy J, Beltrand J, Polak M, Vilsbøll T, Greeley SA, Hattersley AT, Tuomi T. Systematic Review of Treatment of Beta-Cell Monogenic Diabetes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.12.23289807. [PMID: 37214872 PMCID: PMC10197799 DOI: 10.1101/2023.05.12.23289807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Background Beta-cell monogenic forms of diabetes are the area of diabetes care with the strongest support for precision medicine. We reviewed treatment of hyperglycemia in GCK-related hyperglycemia, HNF1A-HNF4A- and HNF1B-diabetes, Mitochondrial diabetes (MD) due to m.3243A>G variant, 6q24-transient neonatal diabetes (TND) and SLC19A2-diabetes. Methods Systematic reviews with data from PubMed, MEDLINE and Embase were performed for the different subtypes. Individual and group level data was extracted for glycemic outcomes in individuals with genetically confirmed monogenic diabetes. Results 147 studies met inclusion criteria with only six experimental studies and the rest being single case reports or cohort studies. Most studies had moderate or serious risk of bias.For GCK-related hyperglycemia, six studies (N=35) showed no deterioration in HbA1c on discontinuing glucose lowering therapy. A randomized trial (n=18 per group) showed that sulfonylureas (SU) were more effective in HNF1A-diabetes than in type 2 diabetes, and cohort and case studies supported SU effectiveness in lowering HbA1c. Two crossover trials (n=15 and n=16) suggested glinides and GLP-1 receptor agonists might be used in place of SU. Evidence for HNF4A-diabetes was limited. While some patients with HNF1B-diabetes (n=301) and MD (n=250) were treated with oral agents, most were on insulin. There was some support for the use of oral agents after relapse in 6q24-TND, and for thiamine improving glycemic control and reducing insulin requirement in SLC19A2-diabetes (less than half achieved insulin-independency). Conclusion There is limited evidence to guide the treatment in monogenic diabetes with most studies being non-randomized and small. The data supports: no treatment in GCK-related hyperglycemia; SU for HNF1A-diabetes. Further evidence is needed to examine the optimum treatment in monogenic subtypes.
Collapse
Affiliation(s)
- Rochelle N. Naylor
- Departments of Pediatrics and Medicine, University of Chicago, Chicago, Illinois, USA
| | - Kashyap A. Patel
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Exeter, Devon, UK
| | - Jarno L.T. Kettunen
- Helsinki University Hospital, Abdominal Centre/Endocrinology, Helsinki, Finland; Folkhalsan Research Center, Helsinki, Finland; Institute for Molecular Medicine Finland FIMM, University of Helsinki, Helsinki, Finland
| | - Jonna M.E. Männistö
- Departments of Pediatrics and Clinical Genetics, Kuopio University Hospital, Kuopio, Finland; Department of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Julie Støy
- Steno diabetes center Aarhus, Aarhus university hospital, Aarhus, Denmark
| | - Jacques Beltrand
- APHP Centre Hôpital Necker Enfants Malades Université Paris Cité, Paris France; Inserm U1016 Institut Cochin Paris France
| | - Michel Polak
- Department of pediatric endocrinology gynecology and diabetology, Hôpital Universitaire Necker Enfants Malades, IMAGINE institute, INSERM U1016, Paris, France; Université Paris Cité, Paris, France
| | - ADA/EASD PMDI
- American Diabetes Association/European Association for the Study of Diabetes Precision Medicine Initiative
| | - Tina Vilsbøll
- Department of Clinical Medicine, University of Copenhagen
| | - Siri A.W. Greeley
- Departments of Pediatrics and Medicine, University of Chicago, Chicago, Illinois, USA
| | - Andrew T. Hattersley
- University of Exeter Medical School, Department of Clinical and Biomedical Sciences, Exeter, Devon, UK
| | | |
Collapse
|
27
|
Wang Y, Yu H, Wang L, Hu J, Feng J. Progress in the preparation and evaluation of glucose-sensitive microneedle systems and their blood glucose regulation. Biomater Sci 2023; 11:5410-5438. [PMID: 37395463 DOI: 10.1039/d3bm00463e] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Glucose-sensitive microneedle systems (GSMSs) as an intelligent strategy for treating diabetes can well solve the problems of puncture pain, hypoglycemia, skin damage, and complications caused by the subcutaneous injection of insulin. According to the various functions of each component, herein, therapeutic GSMSs are reviewed based on three parts (glucose-sensitive models, diabetes medications, and microneedle body). Moreover, the characteristics, benefits, and drawbacks of three types of typical glucose-sensitive models (phenylboronic acid based polymer, glucose oxidase, and concanavalin A) and their drug delivery models are reviewed. In particular, phenylboronic acid-based GSMSs can provide a long-acting drug dose and controlled release rate for the treatment of diabetes. Moreover, their painless, minimally invasive puncture also greatly improves patient compliance, treatment safety, and potential application prospects.
Collapse
Affiliation(s)
- Yu Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, P. R. China.
| | - Haojie Yu
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, P. R. China.
- Zhejiang-Russia Joint Laboratory of Photo-Electro-Magnetic Functional Materials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, P. R. China
| | - Li Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, P. R. China.
- Zhejiang-Russia Joint Laboratory of Photo-Electro-Magnetic Functional Materials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, P. R. China
| | - Jian Hu
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang Province, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, P.R. China
| | - Jingyi Feng
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang Province, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, P.R. China
| |
Collapse
|
28
|
Kilpatrick ES. New Laboratory Guidelines for Diabetes: Continuing the Collaboration between Clinical and Laboratory Medicine. Clin Chem 2023:hvad092. [PMID: 37473456 DOI: 10.1093/clinchem/hvad092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 07/22/2023]
Affiliation(s)
- Eric S Kilpatrick
- Division of Clinical Biochemistry, Sidra Medicine, Doha, Qatar and Weill Cornell Medicine-Qatar, Doha, Qatar
| |
Collapse
|
29
|
Carpenter-Clawson C, Watson M, Pope A, Lynch K, Miles T, Bell D, Talbot M, Varadi A. Competencies of the UK nursing and midwifery workforce to mainstream genomics in the National Health Service: the ongoing gap between perceived importance and confidence in genomics. Front Genet 2023; 14:1125599. [PMID: 37396047 PMCID: PMC10312078 DOI: 10.3389/fgene.2023.1125599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
The United Kingdom is recognised worldwide as a leader in genomics. The use of genomic technologies in the National Health Service (NHS) is expected to deliver faster and more accurate diagnoses, supporting personalized treatments to improve patient outcomes. The ambition of embedding genomic medicine in the diagnostic pathway requires involvement of the front-line clinical workforce, known as 'mainstreaming'. Nurses and midwives are the largest professionally qualified workforce in the National Health Service thus, it is anticipated that they will play key roles in mainstreaming. This study investigated the level of competence/confidence of practicing nurses and midwives to support mainstreaming and their perception of the importance of genomics in delivery of patient care. A literature review of genetics/genomics competency frameworks, semi structured interviews of lead nurses and stakeholders were conducted to identify relevant competencies needed for mainstreaming. These were then used to survey four cohorts of nurses (n = 153) across England in four consecutive years (2019-22). The confidence level of these professionals in all aspects of genomics was 2.07 ± 0.47 measured on a 5-point Likert scale (1"Low confidence"; 5 "High confidence"). Intriguingly, these professionals all appreciated the importance of genomics for their patient care (4.01 ± 0.06). Whilst the importance scores increased, the confidence scores declined at the time when major genomic transformation took place in the NHS (e.g.: launch of the Genomic Medicine Service, the National Genomic Test Directory). To bridge this gap, relevant genomic education can play key roles. However, nurses and midwives were found to be grossly underrepresented in formal genomic education courses offered by Health Education England Genomics Education Programme since 2014. This may result from the lack of direct applicability of the currently offered courses for their practice and role. Thematic analysis revealed that nurses and midwives wish to support their patients by providing more information on their condition, inheritance, and treatment options in combination with the use of relevant genetic counselling skills. This study identified easy to follow competencies for embedding genomics into routine clinical care. We propose a training programme that addresses the gap that nurses and midwives currently have, to enable them to harness genomic opportunities for patients and services.
Collapse
Affiliation(s)
- Catherine Carpenter-Clawson
- Bristol Royal Hospital for Children, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Melanie Watson
- South West Genomic Laboratory Hub, North Bristol NHS Trust, Southmead Hospital, Bristol, United Kingdom
| | - Alison Pope
- Genomics Education Programme, Health Education England, Birmingham, United Kingdom
| | - Kathleen Lynch
- Genomics Education Programme, Health Education England, Birmingham, United Kingdom
| | - Tracie Miles
- NHS Southwest Genomic Medicine Service Alliance, Royal Devon and Exeter Foundation Trust, Exeter, United Kingdom
| | - Dany Bell
- Macmillan Cancer Support, London, United Kingdom
| | | | - Aniko Varadi
- Centre for Research in Biosciences, School of Applied Sciences, University of the West of England, Bristol, United Kingdom
| |
Collapse
|
30
|
Kwak SH, Srinivasan S, Chen L, Todd J, Mercader J, Jensen E, Divers J, Mottl A, Pihoker C, Gandica R, Laffel L, Isganaitis E, Haymond M, Levitsky L, Pollin T, Florez J, Flannick J. Insights from rare variants into the genetic architecture and biology of youth-onset type 2 diabetes. RESEARCH SQUARE 2023:rs.3.rs-2886343. [PMID: 37292813 PMCID: PMC10246295 DOI: 10.21203/rs.3.rs-2886343/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Youth-onset type 2 diabetes (T2D) is a growing public health concern. Its genetic basis and relationship to other forms of diabetes are largely unknown. To gain insight into the genetic architecture and biology of youth-onset T2D, we analyzed exome sequences of 3,005 youth-onset T2D cases and 9,777 ancestry matched adult controls. We identified (a) monogenic diabetes variants in 2.1% of individuals; (b) two exome-wide significant (P < 4.3×10-7) common coding variant associations (in WFS1 and SLC30A8); (c) three exome-wide significant (P < 2.5×10-6) rare variant gene-level associations (HNF1A, MC4R, ATX2NL); and (d) rare variant association enrichments within 25 gene sets broadly related to obesity, monogenic diabetes, and β-cell function. Many association signals were shared between youth-onset and adult-onset T2D but had larger effects for youth-onset T2D risk (1.18-fold increase for common variants and 2.86-fold increase for rare variants). Both common and rare variant associations contributed more to youth-onset T2D liability variance than they did to adult-onset T2D, but the relative increase was larger for rare variant associations (5.0-fold) than for common variant associations (3.4-fold). Youth-onset T2D cases showed phenotypic differences depending on whether their genetic risk was driven by common variants (primarily related to insulin resistance) or rare variants (primarily related to β-cell dysfunction). These data paint a picture of youth-onset T2D as a disease genetically similar to both monogenic diabetes and adult-onset T2D, in which genetic heterogeneity might be used to sub-classify patients for different treatment strategies.
Collapse
Affiliation(s)
| | | | - Ling Chen
- Diabetes Research Center (Diabetes Unit), Department of Medicine, Massachusetts General Hospital
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Jason Flannick
- Broad Institute of MIT and Harvard/Boston Children's Hospital/Harvard Medical School
| |
Collapse
|
31
|
Tan WX, Sim X, Khoo CM, Teo AKK. Prioritization of genes associated with type 2 diabetes mellitus for functional studies. Nat Rev Endocrinol 2023:10.1038/s41574-023-00836-1. [PMID: 37169822 DOI: 10.1038/s41574-023-00836-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/28/2023] [Indexed: 05/13/2023]
Abstract
Existing therapies for type 2 diabetes mellitus (T2DM) show limited efficacy or have adverse effects. Numerous genetic variants associated with T2DM have been identified, but progress in translating these findings into potential drug targets has been limited. Here, we describe the tools and platforms available to identify effector genes from T2DM-associated coding and non-coding variants and prioritize them for functional studies. We discuss QSER1 and SLC12A8 as examples of genes that have been identified as possible T2DM candidate genes using these tools and platforms. We suggest further approaches, including the use of sequencing data with increased sample size and ethnic diversity, single-cell omics data for analyses, glycaemic trait associations to predict gene function and, potentially, human induced pluripotent stem cell 'village' cultures, to strengthen current gene functionalization workflows. Effective prioritization of T2DM-associated genes for experimental validation could expedite our understanding of the genetic mechanisms responsible for T2DM to facilitate the use of precision medicine in its treatment.
Collapse
Affiliation(s)
- Wei Xuan Tan
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Chin Meng Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Adrian K K Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Precision Medicine Translational Research Programme, 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
|
32
|
Aarthy R, Aston-Mourney K, Amutha A, Mikocka-Walus A, Anjana RM, Unnikrishnan R, Jebarani S, Venkatesan U, Gopi S, Radha V, Mohan V. Prevalence, clinical features and complications of common forms of Maturity Onset Diabetes of the Young (MODY) seen at a tertiary diabetes centre in south India. Prim Care Diabetes 2023:S1751-9918(23)00071-2. [PMID: 37055265 DOI: 10.1016/j.pcd.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 03/17/2023] [Accepted: 04/07/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND Maturity Onset Diabetes of the Young (MODY) is a form of monogenic diabetes caused by mutations in single genes, affecting adolescents or young adults. MODY is frequently misdiagnosed as type 1 diabetes (T1). Though several studies from India have reported on the genetic aspects of MODY, the clinical profile, complications and treatments given have not been reported so far, nor compared with T1D and type 2 diabetes (T2D). AIM To determine the prevalence, clinical features, and complications of common forms of genetically proven MODY seen at a tertiary diabetes centre in South India and compare them with matched individuals with T1D and T2D. METHODS Five hundred and thirty individuals identified as 'possible MODY' based on clinical criteria, underwent genetic testing for MODY. Diagnosis of MODY was confirmed based on pathogenic or likely pathogenic variants found using Genome Aggregation Database (gnomAD) and American College of Medical Genetics (ACMG) criteria. The clinical profile of MODY was compared with individuals with type 1 (T1D) and type 2 (T2D) diabetes, matched for duration of diabetes. Retinopathy was diagnosed by retinal photography; nephropathy by urinary albumin excretion > 30 µg/mg of creatinine and neuropathy by vibration perception threshold > 20 v on biothesiometry. RESULTS Fifty-eight patients were confirmed to have MODY (10.9%). HNF1A-MODY (n = 25) was the most common subtype followed by HNF4A-MODY (n = 11), ABCC8-MODY (n = 11), GCK-MODY (n = 6) and HNF1B-MODY (n = 5). For comparison of clinical profile, only the three 'actionable' subtypes - defined as those who may respond to sulphonylureas, namely, HNF1A, HNF4A and ABCC8-MODY, were included. Age at onset of diabetes was lower among HNF4A-MODY and HNF1A-MODY than ABCC8-MODY, T1D and T2D. Prevalence of retinopathy and nephropathy was higher among the three MODY subtypes taken together (n = 47) as compared to T1D (n = 86) and T2D (n = 86). CONCLUSION This is one of the first reports of MODY subtypes from India based on ACMG and gnomAD criteria. The high prevalence of retinopathy and nephropathy in MODY points to the need for earlier diagnosis and better control of diabetes in individuals with MODY.
Collapse
Affiliation(s)
- Ramasamy Aarthy
- Madras Diabetes Research Foundation, Chennai, India; Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Australia
| | - Kathryn Aston-Mourney
- Deakin University, School of Medicine, IMPACT, Institute for Innovation in Physical and Mental Health and Clinical Translation, Geelong, Australia
| | | | | | - Ranjit Mohan Anjana
- Madras Diabetes Research Foundation, Chennai, India; Dr. Mohan's Diabetes Specialties Centre, Chennai, India
| | - Ranjit Unnikrishnan
- Madras Diabetes Research Foundation, Chennai, India; Dr. Mohan's Diabetes Specialties Centre, Chennai, India
| | | | | | | | | | - Viswanathan Mohan
- Madras Diabetes Research Foundation, Chennai, India; Dr. Mohan's Diabetes Specialties Centre, Chennai, India.
| |
Collapse
|
33
|
Şıklar Z, Kontbay T, Colclough K, Patel KA, Berberoğlu M. Expanding the Phenotype of TRMT10A Mutations: Case Report and a Review of the Existing Cases. J Clin Res Pediatr Endocrinol 2023; 15:90-96. [PMID: 34541035 PMCID: PMC9976169 DOI: 10.4274/jcrpe.galenos.2021.2021.0110] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
The tRNA methyltransferase 10 homologue A (TRMT10A) gene encodes tRNA methyl transferase, and biallelic loss of function mutations cause a recessive syndrome of intellectual disability, microcephaly, short stature and diabetes. A case with intellectual disability and distinctive features including microcephaly was admitted. She was diagnosed with epilepsy at 2.5 years old. At 3.6 years of age, severe short stature related to growth hormone (GH) deficiency was detected. She had an incidental diagnosis of diabetes at age 11.4 years which was negative for diabetes antibodies with persistent C-peptide level and she was treated with metformin. Spontaneous puberty did not begin until 15.7 years of age and she was found to have primary ovarian failure. A homozygous p.Arg127* mutation in TRMT10A was detected. In addition to the typical clinical features which characterize TRMT10A syndrome, we observed an unusual form of impaired glucose metabolism which presented in early childhood with hypoglycemia followed by diabetes in late childhood. GH deficiency and primary ovarian failure may also be additional findings of this syndrome. Patients with slow onset diabetes who are negative for auto-antibodies and have extra-pancreatic features should be tested for all known subtypes of monogenic diabetes.
Collapse
Affiliation(s)
- Zeynep Şıklar
- Ankara University Faculty of Medicine, Department of Pediatric Endocrinology, Ankara, Turkey,* Address for Correspondence: Ankara University Faculty of Medicine, Department of Pediatric Endocrinology, Ankara, Turkey Phone: +90 505 342 21 69 E-mail:
| | - Tuğba Kontbay
- Ankara University Faculty of Medicine, Department of Pediatric Endocrinology, Ankara, Turkey
| | - Kevin Colclough
- Exeter Genomics Laboratory, Royal Devon and Exeter Hospital, Exeter, UK
| | - Kashyap A. Patel
- University of Exeter, College of Medicine and Health, Exeter, UK
| | - Merih Berberoğlu
- Ankara University Faculty of Medicine, Department of Pediatric Endocrinology, Ankara, Turkey
| |
Collapse
|
34
|
Arosemena M, Salguero MV, Naylor RN, Wroblewski K, Tasali E, Philipson LH. Objective and Subjective Sleep Patterns in Adults With Maturity-Onset Diabetes of the Young (MODY). Diabetes Care 2023; 46:608-612. [PMID: 36637968 PMCID: PMC10090264 DOI: 10.2337/dc22-1343] [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: 07/09/2022] [Accepted: 12/15/2022] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To examine sleep patterns in adults with maturity-onset diabetes of the young (MODY). RESEARCH DESIGN AND METHODS Adults with glucokinase (GCK)-MODY and transcription factor (TF)-related MODY (HNF1A, HNF1B, HNF4A) were recruited (n = 24; age 46.0 years, 79% women, BMI 24.7 kg/m2) from The University of Chicago's Monogenic Diabetes Registry. Sleep patterns were assessed by 2-week wrist actigraphy (total 315 nights), one night of a home sleep apnea test, and validated surveys. RESULTS Overall, compared with established criteria, 29% of participants had sleep latency ≥15 min, 38% had sleep efficiency ≤85%, 46% had wake after sleep onset >40 min, all indicating poor objective sleep quality. Among all participants, 54% had a sleep duration below the recommended minimum of 7 h, 88% reported poor sleep quality, 58% had obstructive sleep apnea, and 71% reported insomnia. Compared with GCK-MODY, participants with TF-related MODY had poorer objective sleep quality and increased night-to-night variability in sleep patterns. CONCLUSIONS Sleep disturbances appear to be common in adults with MODY despite absent traditional risk factors for sleep disorders. Future research investigating the sleep-diabetes relationship is warranted in this population.
Collapse
Affiliation(s)
| | - Maria V. Salguero
- Department of Medicine, The University of Chicago, Chicago, IL
- Department of Pediatrics, The University of Chicago, Chicago, IL
| | - Rochelle N. Naylor
- Department of Medicine, The University of Chicago, Chicago, IL
- Department of Pediatrics, The University of Chicago, Chicago, IL
| | - Kristen Wroblewski
- Department of Public Health Sciences, The University of Chicago, Chicago, IL
| | - Esra Tasali
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Louis H. Philipson
- Department of Medicine, The University of Chicago, Chicago, IL
- Department of Pediatrics, The University of Chicago, Chicago, IL
| |
Collapse
|
35
|
The Pathogenic Diagnosis in Pediatric Diabetology: Next Generation Sequencing and Precision Therapy. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020310. [PMID: 36837511 PMCID: PMC9964636 DOI: 10.3390/medicina59020310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023]
Abstract
In pediatric diabetology, a precise diagnosis is very important because it allows early and correct clinical management of the patient. Monogenic diabetes (MD), which accounts for 1-6% of all pediatric-adolescent diabetes cases, is the most relevant example of precision medicine. The definitive diagnosis of MD, possible only by genetic testing, allows us to direct patients to more appropriate therapy in relation to the identified mutation. In some cases, MD patients can avoid insulin and be treated with oral hypoglycemic drugs with a perceptible impact on both the quality of life and the healthcare costs. However, the genetic and phenotypic heterogeneity of MD and the overlapping clinical characteristics between different forms, can complicate the diagnostic process. In recent years, the development of Next-Generation Sequencing (NGS) methodology, which allows the simultaneous analysis of multiple genes, has revolutionized molecular diagnostics, becoming the cornerstone of MD precision diagnosis. We report two cases of patients with clinical suspects of MD in which a genetic test was carried out, using a NGS multigenic panel, and it clarified the correct pathogenesis of diabetes, allowing us to better manage the disease both in probands and other affected family members.
Collapse
|
36
|
Watanabe H, Goto S, Hosojima M, Kabasawa H, Imai N, Ito Y, Narita I. Pathogenic variants of Alport syndrome and monogenic diabetes identified by exome sequencing in a family. Hum Genome Var 2023; 10:5. [PMID: 36732323 PMCID: PMC9894847 DOI: 10.1038/s41439-023-00233-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 02/04/2023] Open
Abstract
We present a family of two female Alport syndrome patients with a family history of impaired glucose tolerance. Whole exome sequencing identified a novel heterozygous variant of COL4A5 NM_033380.3: c.2636 C > A (p.S879*) and a rare variant of GCK NM_001354800.1: c.1135 G > A (p.A379T) as the causes of Alport syndrome and monogenic diabetes, respectively. Two independent pathogenic variants affected the clinical phenotypes. Clinical next-generation sequencing is helpful for identifying the causes of patients' manifestations.
Collapse
Affiliation(s)
- Hirofumi Watanabe
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| | - Shin Goto
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Michihiro Hosojima
- Department of Clinical Nutrition Science, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hideyuki Kabasawa
- Department of Clinical Nutrition Science, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Naofumi Imai
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yumi Ito
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Department of Health Promotion Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
37
|
Shields BM, Dennis JM, Angwin CD, Warren F, Henley WE, Farmer AJ, Sattar N, Holman RR, Jones AG, Pearson ER, Hattersley AT. Patient stratification for determining optimal second-line and third-line therapy for type 2 diabetes: the TriMaster study. Nat Med 2023; 29:376-383. [PMID: 36477733 PMCID: PMC7614216 DOI: 10.1038/s41591-022-02120-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/07/2022] [Indexed: 12/13/2022]
Abstract
Precision medicine aims to treat an individual based on their clinical characteristics. A differential drug response, critical to using these features for therapy selection, has never been examined directly in type 2 diabetes. In this study, we tested two hypotheses: (1) individuals with body mass index (BMI) > 30 kg/m2, compared to BMI ≤ 30 kg/m2, have greater glucose lowering with thiazolidinediones than with DPP4 inhibitors, and (2) individuals with estimated glomerular filtration rate (eGFR) 60-90 ml/min/1.73 m2, compared to eGFR >90 ml/min/1.73 m2, have greater glucose lowering with DPP4 inhibitors than with SGLT2 inhibitors. The primary endpoint for both hypotheses was the achieved HbA1c difference between strata for the two drugs. In total, 525 people with type 2 diabetes participated in this UK-based randomized, double-blind, three-way crossover trial of 16 weeks of treatment with each of sitagliptin 100 mg once daily, canagliflozin 100 mg once daily and pioglitazone 30 mg once daily added to metformin alone or metformin plus sulfonylurea. Overall, the achieved HbA1c was similar for the three drugs: pioglitazone 59.6 mmol/mol, sitagliptin 60.0 mmol/mol and canagliflozin 60.6 mmol/mol (P = 0.2). Participants with BMI > 30 kg/m2, compared to BMI ≤ 30 kg/m2, had a 2.88 mmol/mol (95% confidence interval (CI): 0.98, 4.79) lower HbA1c on pioglitazone than on sitagliptin (n = 356, P = 0.003). Participants with eGFR 60-90 ml/min/1.73 m2, compared to eGFR >90 ml/min/1.73 m2, had a 2.90 mmol/mol (95% CI: 1.19, 4.61) lower HbA1c on sitagliptin than on canagliflozin (n = 342, P = 0.001). There were 2,201 adverse events reported, and 447/525 (85%) randomized participants experienced an adverse event on at least one of the study drugs. In this precision medicine trial in type 2 diabetes, our findings support the use of simple, routinely available clinical measures to identify the drug class most likely to deliver the greatest glycemic reduction for a given patient. (ClinicalTrials.gov registration: NCT02653209 ; ISRCTN registration: 12039221 .).
Collapse
Affiliation(s)
- Beverley M Shields
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - John M Dennis
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - Catherine D Angwin
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - Fiona Warren
- Clinical Trials Unit, University of Exeter Medical School, Exeter, UK
- Institute of Health Research, University of Exeter Medical School, Exeter, UK
| | - William E Henley
- Institute of Health Research, University of Exeter Medical School, Exeter, UK
| | - Andrew J Farmer
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Naveed Sattar
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, UK
| | - Rury R Holman
- Diabetes Trials Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Angus G Jones
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - Ewan R Pearson
- Population Health & Genomics, School of Medicine, University of Dundee, Dundee, UK
| | - Andrew T Hattersley
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK.
| | | |
Collapse
|
38
|
Chow E, Wang K, Lim CK, Tsoi ST, Fan B, Poon E, Luk AO, Ma RC, Ferrannini E, Mari A, Chen L, Chan JC. Dorzagliatin, a Dual-Acting Glucokinase Activator, Increases Insulin Secretion and Glucose Sensitivity in Glucokinase Maturity-Onset Diabetes of the Young and Recent-Onset Type 2 Diabetes. Diabetes 2023; 72:299-308. [PMID: 36342518 PMCID: PMC9871194 DOI: 10.2337/db22-0708] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
Glucokinase (GK, gene symbol GCK) maturity-onset diabetes of the young (MODY) is caused by heterozygous inactivating mutations in GK and impaired glucose sensing. We investigated effects of dorzagliatin, a novel allosteric GK activator, on insulin secretion rates (ISRs) and β-cell glucose sensitivity (βCGS) in GCK-MODY and recent-onset type 2 diabetes. In a double-blind, randomized, crossover study, 8 participants with GCK-MODY and 10 participants with type 2 diabetes underwent 2-h 12 mmol/L hyperglycemic clamps following a single oral dose of dorzagliatin 75 mg or matched placebo. Effects of dorzagliatin on wild-type and mutant GK enzyme activity were investigated using an NADP+-coupled assay with glucose-6-phosphate dehydrogenase in vitro. In GCK-MODY, dorzagliatin significantly increased absolute and incremental second-phase ISRs versus placebo but not the acute insulin response. Dorzagliatin improved βCGS in GCK-MODY with an upward and leftward shift in ISR-glucose response. Dorzagliatin increased basal ISRs in type 2 diabetes, with smaller changes in second-phase ISRs versus GCK-MODY. In vitro, dorzagliatin directly reduced the glucose half saturation concentration of wild-type GK and selected GK mutants to varying degrees. Dorzagliatin directly restored enzyme activity of select GK mutants and enhanced wild-type GK activity, thereby correcting the primary defect of glucose sensing in GCK-MODY.
Collapse
Affiliation(s)
- Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
- Corresponding authors: Juliana C.N. Chan, , and Elaine Chow,
| | - Ke Wang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Cadmon K.P. Lim
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong, China
| | - Sandra T.F. Tsoi
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong, China
| | - Baoqi Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong, China
| | - Emily Poon
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Andrea O.Y. Luk
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Ronald C.W. Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong, China
| | - Ele Ferrannini
- Institute of Clinical Physiology, National Research Council of Italy (CNR), Pisa, Italy
| | - Andrea Mari
- Institute of Neuroscience, National Research Council of Italy (CNR), Padua, Italy
| | - Li Chen
- Hua Medicine, Shanghai, China
| | - Juliana C.N. Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong, China
- Corresponding authors: Juliana C.N. Chan, , and Elaine Chow,
| |
Collapse
|
39
|
Chen Y, Wang Q, Xie Z, Huang G, Fan L, Li X, Zhou Z. The impact of family history of type 2 diabetes on clinical heterogeneity in idiopathic type 1 diabetes. Diabetes Obes Metab 2023; 25:417-425. [PMID: 36200314 DOI: 10.1111/dom.14884] [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: 08/09/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 02/02/2023]
Abstract
AIM To investigate the impact of family history of type 2 diabetes (T2D) on the clinical phenotypes of patients with idiopathic type 1 diabetes (T1D). METHODS In clinically diagnosed T1D cases, a total of 335 idopathic T1D patients were included in the study, after excluding autoimmune T1D using islet autoantibody testing and monogenic diabetes using a custom monogenic diabetes gene panel obtained from clinically diagnosed T1D cases. A semi-structured questionnaire was used to collect information on the presence of T2D in first-degree relatives. The demographic and metabolic markers of idiopathic T1D patients were analysed. Subgroup analysis was performed to investigate potential interactions between T2D family history and human leukocyte antigen (HLA) genotypes. RESULTS A total of 18.2% of individuals with idiopathic T1D had a T2D family history, and these individuals were more likely to have features associated with T2D, such as older age of onset, higher body mass index at diagnosis, lower insulin dosage and better beta-cell function, as indicated by higher levels of fasting C-peptide and 2-hour postprandial C-peptide (all P < 0.05). Additionally, regardless of HLA susceptible genotypes, the impact of family history of T2D was consistently observed in idiopathic T1D patients. Multivariable analyses showed that T2D family history was negatively correlated with the risk of beta-cell function failure in idiopathic T1D patients (P < 0.05). CONCLUSIONS Family history of T2D may be implicated in the heterogeneity of idiopathic T1D patients.
Collapse
Affiliation(s)
- Yan Chen
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qianrong Wang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhiguo Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Gan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Li Fan
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
40
|
Liu J, Fu J, Xie Z, Ding L, Wang D, Yu M, Zhang Q, Xie T, Xiao X. Serum metabolomics identified metabolite biomarkers and distinguished maturity-onset diabetes of the young from type 1 diabetes in the Chinese population. Clin Chim Acta 2023; 539:250-258. [PMID: 36584766 DOI: 10.1016/j.cca.2022.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Maturity-onset diabetes of the young (MODY) patients have unique clinical manifestations and need individualized treatments. We identified novel serum metabolic biomarkers to distinguish MODY and explore the possible mechanism of the clinical manifestation and complications of MODY. METHODS Fasting serum samples were collected from MODY3 (n = 17), MODY2 (n = 33), type 1 diabetes (T1DM) (n = 34) and healthy individuals (n = 30), and were analyzed using the ultra-performance liquid chromatography-mass spectrometry (UPLC-MS) metabolomic platform. RESULTS 4 metabolites were found significantly fluctuated between groups, including glycerophosphocholine, LysoPC(18:2(9Z,12Z)), sphinganine and l-Phenylalanine. Glycerophosphocholine was selected as a diagnostic biomarker. The the area under the ROC curve (AUC) for distinguishing MODYs from healthy controls and differentiating MODY3 from T1DM reached 1.0. The combination of metabolites also gained good diagnostic value. The AUC of the combination of LysoPC(18:2(9Z,12Z)), sphinganine and l-Phenylalanine for discriminating MODY3 from T1DM was 0.983. Besides, the combination of clinical indices and metabolites helped to better differentiate the 2 MODY subtypes. CONCLUSIONS We identified the metabolic profiles of MODY2 and MODY3 and found promising biomarkers for distinguishing MODY from T1DM, which provides evidence for the pathogenesis and characteristic clinical manifestations of patients with MODY2 and MODY3.
Collapse
Affiliation(s)
- Jieying Liu
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Junling Fu
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; Department of Endocrinology, Beijing Institute of Geriatrics, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Ziyan Xie
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Lu Ding
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Dongmei Wang
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Miao Yu
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Qian Zhang
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Ting Xie
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China; Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Xinhua Xiao
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
41
|
Moreno Tirado A, Del Val Zaballos F. Neonatal diabetes with response to very low doses of sulfonylureas after 47 years of diagnosis. Med Clin (Barc) 2023; 160:370-371. [PMID: 36642651 DOI: 10.1016/j.medcli.2022.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 01/15/2023]
|
42
|
Chen Y, Zhao J, Li X, Xie Z, Huang G, Yan X, Zhou H, Zheng L, Xu T, Zhou K, Zhou Z. Prevalence of maturity-onset diabetes of the young in phenotypic type 2 diabetes in young adults: a nationwide, multi-center, cross-sectional survey in China. Chin Med J (Engl) 2023; 136:56-64. [PMID: 36723869 PMCID: PMC10106210 DOI: 10.1097/cm9.0000000000002321] [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: 04/29/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Maturity-onset diabetes of the young (MODY) is the most common monogenic diabetes. The aim of this study was to assess the prevalence of MODY in phenotypic type 2 diabetes (T2DM) among Chinese young adults. METHODS From April 2015 to October 2017, this cross-sectional study involved 2429 consecutive patients from 46 hospitals in China, newly diagnosed between 15 years and 45 years, with T2DM phenotype and negative for standardized glutamic acid decarboxylase antibody at the core laboratory. Sequencing using a custom monogenic diabetes gene panel was performed, and variants of 14 MODY genes were interpreted as per current guidelines. RESULTS The survey determined 18 patients having genetic variants causing MODY (6 HNF1A , 5 GCK , 3 HNF4A , 2 INS , 1 PDX1 , and 1 PAX4 ). The prevalence of MODY was 0.74% (95% confidence interval [CI]: 0.40-1.08%). The clinical characteristics of MODY patients were not specific, 72.2% (13/18) of them were diagnosed after 35 years, 47.1% (8/17) had metabolic syndrome, and only 38.9% (7/18) had a family history of diabetes. No significant difference in manifestations except for hemoglobin A1c levels was found between MODY and non-MODY patients. CONCLUSION The prevalence of MODY in young adults with phenotypic T2DM was 0.74%, among which HNF1A -, GCK -, and HNF4A -MODY were the most common subtypes. Clinical features played a limited role in the recognition of MODY.
Collapse
Affiliation(s)
- Yan Chen
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Jing Zhao
- College of Life Sciences, The University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhiguo Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Gan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xiang Yan
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Houde Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Li Zheng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Tao Xu
- College of Life Sciences, The University of Chinese Academy of Sciences, Beijing 100049, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 271016, China
| | - Kaixin Zhou
- College of Life Sciences, The University of Chinese Academy of Sciences, Beijing 100049, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 271016, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
43
|
Kaser S, Hofer SE, Kazemi-Shirazi L, Festa A, Winhofer Y, Sourij H, Brath H, Riedl M, Resl M, Clodi M, Stulnig T, Ress C, Luger A. [Other specific types of diabetes and exocrine pancreatic insufficiency (update 2023)]. Wien Klin Wochenschr 2023; 135:18-31. [PMID: 37101022 PMCID: PMC10133035 DOI: 10.1007/s00508-022-02123-x] [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] [Accepted: 11/09/2022] [Indexed: 04/28/2023]
Abstract
The heterogenous category "specific types of diabetes due to other causes" encompasses disturbances in glucose metabolism due to other endocrine disorders such as acromegaly or hypercortisolism, drug-induced diabetes (e.g. antipsychotic medications, glucocorticoids, immunosuppressive agents, highly active antiretroviral therapy (HAART), checkpoint inhibitors), genetic forms of diabetes (e.g. Maturity Onset Diabetes of the Young (MODY), neonatal diabetes, Down‑, Klinefelter- and Turner Syndrome), pancreatogenic diabetes (e.g. postoperatively, pancreatitis, pancreatic cancer, haemochromatosis, cystic fibrosis), and some rare autoimmune or infectious forms of diabetes. Diagnosis of specific diabetes types might influence therapeutic considerations. Exocrine pancreatic insufficiency is not only found in patients with pancreatogenic diabetes but is also frequently seen in type 1 and long-standing type 2 diabetes.
Collapse
Affiliation(s)
- Susanne Kaser
- Universitätsklinik für Innere Medizin 1, Medizinische Universität Innsbruck, Anichstraße 35, 6020, Innsbruck, Österreich.
| | - Sabine E Hofer
- Universitätsklinik für Pädiatrie 1, Medizinische Universität Innsbruck, Innsbruck, Österreich
| | - Lili Kazemi-Shirazi
- Klinische Abteilung für Gastroenterologie und Hepatologie, Universitätsklinik für Innere Medizin III, Medizinische Universität Wien, Wien, Österreich
| | - Andreas Festa
- Abteilung für Innere Medizin I, LK Stockerau, Stockerau, Österreich
| | - Yvonne Winhofer
- Klinische Abteilung für Endokrinologie und Stoffwechsel, Universitätsklinik für Innere Medizin III, Medizinische Universität Wien, Wien, Österreich
| | - Harald Sourij
- Klinische Abteilung für Endokrinologie und Diabetologie, Universitätsklinik für Innere Medizin, Medizinische Universität Graz, Graz, Österreich
| | - Helmut Brath
- Mein Gesundheitszentrum Favoriten, Österreichische Gesundheitskasse, Wien, Österreich
| | - Michaela Riedl
- Klinische Abteilung für Endokrinologie und Stoffwechsel, Universitätsklinik für Innere Medizin III, Medizinische Universität Wien, Wien, Österreich
| | - Michael Resl
- Abteilung für Innere Medizin, Konventhospital der Barmherzigen Brüder Linz, Linz, Österreich
| | - Martin Clodi
- Abteilung für Innere Medizin, Konventhospital der Barmherzigen Brüder Linz, Linz, Österreich
- ICMR - Institute for Cardiovascular and Metabolic Research, JKU Linz, Linz, Österreich
| | - Thomas Stulnig
- 3. Medizinische Abteilung und Karl Landsteiner Institut für Stoffwechselerkrankungen und Nephrologie, Klinik Hietzing, Wien, Österreich
| | - Claudia Ress
- Universitätsklinik für Innere Medizin 1, Medizinische Universität Innsbruck, Anichstraße 35, 6020, Innsbruck, Österreich
| | - Anton Luger
- Klinische Abteilung für Endokrinologie und Stoffwechsel, Universitätsklinik für Innere Medizin III, Medizinische Universität Wien, Wien, Österreich
| |
Collapse
|
44
|
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
|
45
|
Mirshahi UL, Colclough K, Wright CF, Wood AR, Beaumont RN, Tyrrell J, Laver TW, Stahl R, Golden A, Goehringer JM, Frayling TF, Hattersley AT, Carey DJ, Weedon MN, Patel KA. Reduced penetrance of MODY-associated HNF1A/HNF4A variants but not GCK variants in clinically unselected cohorts. Am J Hum Genet 2022; 109:2018-2028. [PMID: 36257325 PMCID: PMC9674944 DOI: 10.1016/j.ajhg.2022.09.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/28/2022] [Indexed: 01/26/2023] Open
Abstract
The true prevalence and penetrance of monogenic disease variants are often not known because of clinical-referral ascertainment bias. We comprehensively assess the penetrance and prevalence of pathogenic variants in HNF1A, HNF4A, and GCK that account for >80% of monogenic diabetes. We analyzed clinical and genetic data from 1,742 clinically referred probands, 2,194 family members, clinically unselected individuals from a US health system-based cohort (n = 132,194), and a UK population-based cohort (n = 198,748). We show that one in 1,500 individuals harbor a pathogenic variant in one of these genes. The penetrance of diabetes for HNF1A and HNF4A pathogenic variants was substantially lower in the clinically unselected individuals compared to clinically referred probands and was dependent on the setting (32% in the population, 49% in the health system cohort, 86% in a family member, and 98% in probands for HNF1A). The relative risk of diabetes was similar across the clinically unselected cohorts highlighting the role of environment/other genetic factors. Surprisingly, the penetrance of pathogenic GCK variants was similar across all cohorts (89%-97%). We highlight that pathogenic variants in HNF1A, HNF4A, and GCK are not ultra-rare in the population. For HNF1A and HNF4A, we need to tailor genetic interpretation and counseling based on the setting in which a pathogenic monogenic variant was identified. GCK is an exception with near-complete penetrance in all settings. This along with the clinical implication of diagnosis makes it an excellent candidate for the American College of Medical Genetics secondary gene list.
Collapse
Affiliation(s)
| | - Kevin Colclough
- Molecular Genetics, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Caroline F Wright
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Andrew R Wood
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Robin N Beaumont
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Jessica Tyrrell
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Thomas W Laver
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Richard Stahl
- Geisinger Clinic, Geisinger Health System, Danville, PA, USA
| | - Alicia Golden
- Geisinger Clinic, Geisinger Health System, Danville, PA, USA
| | | | - Timothy F Frayling
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Andrew T Hattersley
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, UK
| | - David J Carey
- Geisinger Clinic, Geisinger Health System, Danville, PA, USA
| | - Michael N Weedon
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, UK.
| | - Kashyap A Patel
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, UK.
| |
Collapse
|
46
|
Li Z, Li K, Sun Y, Jiang X, Liu J, Li J, Fang L, Li G, Guan Q, Xu C. Mutations in GCK May Lead to MODY2 by Reducing Glycogen Synthesis. Adv Biol (Weinh) 2022; 6:e2200097. [PMID: 35770790 DOI: 10.1002/adbi.202200097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/18/2022] [Indexed: 01/28/2023]
Abstract
Dysfunction of glucokinase (GCK) caused by mutations in the GCK gene is the main cause of maturity-onset diabetes of the young type-2 (MODY2, also known as GCK-MODY), which is usually present in adolescence or young adulthood. MODY2 is characterized by mild, stable fasting hyperglycemia that presents at birth, usually 5.4-8.3 mmol L-1 , and rarely develops complications from diabetes. The treatment of MODY2 prefers a manageable diet rather than the use of insulin. Previous studies have identified GCK mutations only by online software prediction or enzyme kinetic analysis and thermolability assays which are complicated to be conducted. In this study, six mutations in the GCK gene, including four novel mutations and two mutations that are previously reported, are identified. All the six locations are highly conserved according to the sequencing alignment. Moreover, missense mutations are strongly predicted to be pathogenic using online programs. Functional studies show that mutations in GCK mutation do not affect insulin secretion but affect glycogen synthesis. These findings demonstrate that GCK mutations decrease glycogen synthesis, which leads to hyperglycemia in MODY2. Meanwhile, this study provides a new perspective and methods for identifying pathogenic mutations in GCK.
Collapse
Affiliation(s)
- Zongyue Li
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.,Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China.,Shandong Institute of Endocrine and Metabolic Disease, Jinan, Shandong, 250021, China
| | - Kunxia Li
- Department of Pediatric, Yantai Yuhuangding Hospital affiliated to Qingdao University, Qingdao, Shandong, 266000, China
| | - Yan Sun
- Department of Pediatrics, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Xiuyun Jiang
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Jia Liu
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Jingyi Li
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.,Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China.,Shandong Institute of Endocrine and Metabolic Disease, Jinan, Shandong, 250021, China
| | - Li Fang
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China.,Shandong Institute of Endocrine and Metabolic Disease, Jinan, Shandong, 250021, China.,Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Guimei Li
- Department of Pediatrics, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Qingbo Guan
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Chao Xu
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.,Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, China.,Shandong Institute of Endocrine and Metabolic Disease, Jinan, Shandong, 250021, China.,Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| |
Collapse
|
47
|
Stankute I, Dobrovolskiene R, Danyte E, Steponaviciute R, Schwitzgebel VM, Verkauskiene R. Pancreatic beta-cell function dynamics in youth with GCK, HNF1A, and KCNJ11 genes mutations during mixed meal tolerance test. Pediatr Diabetes 2022; 23:1009-1016. [PMID: 36068963 PMCID: PMC9826376 DOI: 10.1111/pedi.13404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/21/2022] [Accepted: 08/14/2022] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE The aims were (1) to assess beta-cell function in GCK diabetes patients over 2-year period; (2) to evaluate the dynamics of beta-cell function in HNF1A and KCNJ11 patients after treatment optimization; using mixed meal tolerance test (MMTT) as a gold standard for non-invasive beta-cell function assessment. RESEARCH DESIGN AND METHODS Twenty-two GCK diabetes patients, 22 healthy subjects, 4 patients with HNF1A and 2 with KCNJ11 were recruited. Firstly, beta-cell function was compared between GCK patients versus controls; the dynamics of beta-cell function were assessed in GCK patients with two MMTTs in 2-year period. Secondly, the change of beta-cell function was evaluated in HNF1A and KCNJ11 patients after successful treatment optimization in 2-year period. RESULTS GCK diabetes patients had lower area under the curve (AUC) of C-peptide (CP), average CP and peak CP compared to controls. Also, higher levels of fasting, average, peak and AUC of glycemia during MMTT were found in GCK patients compared to healthy controls. No significant changes in either CP or glycemia dynamics were observed in GCK diabetes group comparing 1st and 2nd MMTTs. Patients with HNF1A and KCNJ11 diabetes had significantly improved diabetes control 2 years after the treatment was optimized (HbA1c 7.1% vs. 5.9% [54 mmol/mol vs. 41 mmol/mol], respectively, p = 0.028). Higher peak CP and lower HbA1c were found during 2nd MMTT in patients with targeted treatment compared to the 1st MMTT before the treatment change. CONCLUSION In short-term perspective, GCK diabetes group revealed no deterioration of beta-cell function. Individualized treatment in monogenic diabetes showed improved beta-cell function.
Collapse
Affiliation(s)
- Ingrida Stankute
- Institute of EndocrinologyLithuanian University of Health SciencesKaunasLithuania,Medical AcademyLithuanian University of Health SciencesKaunasLithuania
| | | | - Evalda Danyte
- Institute of EndocrinologyLithuanian University of Health SciencesKaunasLithuania
| | - Rasa Steponaviciute
- Department of Laboratory MedicineLithuanian University of Health SciencesKaunasLithuania
| | - Valerie M. Schwitzgebel
- Pediatric Endocrine and Diabetes Unit, Department of Pediatrics, Gynecology and ObstetricsUniversity Hospitals of GenevaGenevaSwitzerland,Diabetes Center of the Faculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Rasa Verkauskiene
- Institute of EndocrinologyLithuanian University of Health SciencesKaunasLithuania
| |
Collapse
|
48
|
Bonnefond A, Semple RK. Achievements, prospects and challenges in precision care for monogenic insulin-deficient and insulin-resistant diabetes. Diabetologia 2022; 65:1782-1795. [PMID: 35618782 PMCID: PMC9522735 DOI: 10.1007/s00125-022-05720-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/01/2022] [Indexed: 01/19/2023]
Abstract
Integration of genomic and other data has begun to stratify type 2 diabetes in prognostically meaningful ways, but this has yet to impact on mainstream diabetes practice. The subgroup of diabetes caused by single gene defects thus provides the best example to date of the vision of 'precision diabetes'. Monogenic diabetes may be divided into primary pancreatic beta cell failure, and primary insulin resistance. In both groups, clear examples of genotype-selective responses to therapy have been advanced. The benign trajectory of diabetes due to pathogenic GCK mutations, and the sulfonylurea-hyperresponsiveness conferred by activating KCNJ11 or ABCC8 mutations, or loss-of-function HNF1A or HNF4A mutations, often decisively guide clinical management. In monogenic insulin-resistant diabetes, subcutaneous leptin therapy is beneficial in some severe lipodystrophy. Increasing evidence also supports use of 'obesity therapies' in lipodystrophic people even without obesity. In beta cell diabetes the main challenge is now implementation of the precision diabetes vision at scale. In monogenic insulin-resistant diabetes genotype-specific benefits are proven in far fewer patients to date, although further genotype-targeted therapies are being evaluated. The conceptual paradigm established by the insulin-resistant subgroup with 'adipose failure' may have a wider influence on precision therapy for common type 2 diabetes, however. For all forms of monogenic diabetes, population-wide genome sequencing is currently forcing reappraisal of the importance assigned to pathogenic mutations when gene sequencing is uncoupled from prior suspicion of monogenic diabetes.
Collapse
Affiliation(s)
- Amélie Bonnefond
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, France.
- Université de Lille, Lille, France.
- Department of Metabolism, Imperial College London, London, UK.
| | - Robert K Semple
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK.
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
49
|
Colclough K, Patel K. How do I diagnose Maturity Onset Diabetes of the Young in my patients? Clin Endocrinol (Oxf) 2022; 97:436-447. [PMID: 35445424 PMCID: PMC9544561 DOI: 10.1111/cen.14744] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/21/2022] [Accepted: 04/13/2022] [Indexed: 11/28/2022]
Abstract
Maturity Onset Diabetes of the Young (MODY) is a monogenic form of diabetes diagnosed in young individuals that lack the typical features of type 1 and type 2 diabetes. The genetic subtype of MODY determines the most effective treatment and this is the driver for MODY genetic testing in diabetes populations. Despite the obvious clinical and health economic benefits, MODY is significantly underdiagnosed with the majority of patients being inappropriately managed as having type 1 or type 2 diabetes. Low detection rates result from the difficulty in identifying patients with a likely diagnosis of MODY from the high background population of young onset type 1 and type 2 diabetes, compounded by the lack of MODY awareness and education in diabetes care physicians. MODY diagnosis can be improved through (1) access to education and training, (2) the use of sensitive and specific selection criteria based on accurate prediction models and biomarkers to identify patients for testing, (3) the development and mainstream implementation of simple criteria-based selection pathways applicable across a range of healthcare settings and ethnicities to select the most appropriate patients for genetic testing and (4) the correct use of next generation sequencing technology to provide accurate and comprehensive testing of all known MODY and monogenic diabetes genes. The creation and public sharing of educational materials, clinical and scientific best practice guidelines and genetic variants will help identify the missing patients so they can benefit from the more effective clinical care that a genetic diagnosis brings.
Collapse
Affiliation(s)
- Kevin Colclough
- Exeter Genomics LaboratoryRoyal Devon & Exeter NHS Foundation TrustExeterUK
| | - Kashyap Patel
- Institute of Biomedical and Clinical ScienceUniversity of Exeter Medical SchoolExeterUK
| |
Collapse
|
50
|
Burahmah J, Zheng D, Leslie RD. Adult-onset type 1 diabetes: A changing perspective. Eur J Intern Med 2022; 104:7-12. [PMID: 35718648 DOI: 10.1016/j.ejim.2022.06.003] [Citation(s) in RCA: 4] [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: 02/01/2022] [Revised: 04/27/2022] [Accepted: 06/03/2022] [Indexed: 11/19/2022]
Abstract
Type 1 diabetes most commonly presents in adulthood, contrary to the widely held view that it is a disease of childhood. Furthermore, a substantial proportion of cases of adult-onset type 1 diabetes does not require insulin therapy at clinical onset. Recent studies have emphasised the evidence that adult-onset type 1 diabetes is prevalent but often misclassified initially as type 2 diabetes (1, 2). In this review, we discuss that recent literature, highlighting the similarities and differences between adult-onset and childhood-onset type 1 diabetes, exploring recent debates surrounding its epidemiology and genetics, as well as expanding on important issues of diagnostic criteria for individuals presenting with adult-onset diabetes and the subsequent management once identified as having an autoimmune basis. In addition, this review looks at the psychosocial challenges faced by T1D patients and their possible management.
Collapse
Affiliation(s)
- J Burahmah
- Blizard Institute, Queen Mary, London, UK
| | - D Zheng
- Blizard Institute, Queen Mary, London, UK
| | - R D Leslie
- Blizard Institute, Queen Mary, London, UK.
| |
Collapse
|