1
|
Kreienkamp RJ, Deutsch AJ, Huerta-Chagoya A, Borglund EM, Florez JC, Flannick J, Udler MS. Type 1 Diabetes Polygenic Scores Improve Diagnostic Accuracy in Pediatric Diabetes Care. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.06.25323229. [PMID: 40162251 PMCID: PMC11952484 DOI: 10.1101/2025.03.06.25323229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Accurately classifying pediatric diabetes can be challenging for providers, and misclassification can result in suboptimal care. In recent years, type 1 diabetes (T1D) polygenic scores, which quantify one's genetic risk for T1D based on T1D risk allele burden, have been developed with good discriminating capacity between T1D and not-T1D. These tools have the potential to improve significantly diagnostic provider accuracy if used in clinic. Methods We applied T1D polygenic scores to a group of pediatric patients (n=1846) with genetic data available in the Boston Children's Hospital PrecisionLink Biobank, including 96 individuals diagnosed with T1D. Results Patients with a clinical diagnosis of T1D had higher T1D polygenic scores compared to controls (Wilcoxon rank-sum P<0.0001). Sixty-nine of the 74 individuals with diabetes and a T1D polygenic score exceeding an externally validated cutoff for distinguishing T1D from not-T1D were confirmed to have T1D. There were multiple cases where T1D polygenic scores would have clinical utility. An elevated T1D polygenic score suggested T1D in a pancreatic autoantibody (PAA)- negative individual with negative MODY genetic testing and a phenotype matching T1D. A low T1D polygenic score accurately indicated atypical diabetes in an individual found to have HNF1B-MODY. One individual had positive PAA, but the provider noted that the patient may not have classic T1D, as later suggested by a low T1D polygenic score. Conclusion T1D polygenic scores already have clinical utility to aid in the accurate diagnosis of pediatric diabetes. Efforts are now needed to advance their use in clinical practice.
Collapse
Affiliation(s)
- Raymond J. Kreienkamp
- Division of Endocrinology, Department of Pediatrics, Boston Children’s Hospital, Boston, 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
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aaron J. Deutsch
- 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
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alicia Huerta-Chagoya
- 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
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Erin M. Borglund
- Computational Health Informatics Program (CHIP), Boston Children’s Hospital, Boston, MA, USA
| | - Jose C. Florez
- 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
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jason Flannick
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Genetics and Genomics, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
| | - Miriam S. Udler
- 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
- Programs in Metabolism and Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
2
|
Tosur M, Onengut-Gumuscu S, Redondo MJ. Type 1 Diabetes Genetic Risk Scores: History, Application and Future Directions. Curr Diab Rep 2025; 25:22. [PMID: 39920466 DOI: 10.1007/s11892-025-01575-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/04/2025] [Indexed: 02/09/2025]
Abstract
PURPOSE OF REVIEW To review the genetics of type 1 diabetes (T1D) and T1D genetic risk scores, focusing on their development, research and clinical applications, and future directions. RECENT FINDINGS More than 90 genetic loci have been linked to T1D risk, with approximately half of the genetic risk attributable to the human leukocyte antigen (HLA) locus, along with non-HLA loci that have smaller effects to disease risk. The practical use of T1D genetic risk scores simplifies the complex genetic information, within the HLA and non-HLA regions, by combining the additive effect and interactions of single nucleotide polymorphisms (SNPs) associated with risk. Genetic risk scores have proven to be useful in various aspects, including classifying diabetes (e.g., distinguishing between T1D vs. neonatal, type 2 or other diabetes types), predicting the risk of developing T1D, assessing the prognosis of the clinical course (e.g., determining the risk of developing insulin dependence and glycemic control), and research into the heterogeneity of diabetes (e.g., atypical diabetes). However, there are gaps in our current knowledge including the specific sets of genes that regulate transition between preclinical stages of T1D, response to disease modifying therapies, and other outcomes of interest such as persistence of beta cell function. Several T1D genetic risk scores have been developed and shown to be valuable in various contexts, from classification of diabetes to providing insights into its etiology and predicting T1D risk across different stages of T1D. Further research is needed to develop and validate T1D genetic risk scores that are effective across all populations and ancestries. Finally, barriers such as cost, and training of medical professionals have to be addressed before the use of genetic risk scores can be incorporated into routine clinical practice.
Collapse
Affiliation(s)
- Mustafa Tosur
- Department of Pediatrics, Division of Diabetes and Endocrinology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA.
- Children's Nutrition Research Center, USDA/ARS, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| | | | - Maria J Redondo
- Department of Pediatrics, Division of Diabetes and Endocrinology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
3
|
Sharma M, Maurya K, Nautiyal A, Chitme HR. Monogenic Diabetes: A Comprehensive Overview and Therapeutic Management of Subtypes of Mody. Endocr Res 2025; 50:1-11. [PMID: 39106207 DOI: 10.1080/07435800.2024.2388606] [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: 02/23/2024] [Revised: 07/21/2024] [Accepted: 07/31/2024] [Indexed: 08/09/2024]
Abstract
BACKGROUND Monogenic diabetes often occurs as a result of single-gene mutations. The illness is minimally affected by environmental and behavioral factors, and it constitutes around one to five percent of all cases of diabetes. METHODS Newborn diabetes mellitus (NDM) and maturity-onset diabetes of the young (MODY) are the predominant causes of monogenic diabetes, accounting for a larger proportion of cases, while syndromic diabetes represents a smaller percentage. MODY, a group of inherited non-autoimmune diabetes mellitus disorders, is quite common. However, it remains frequently misdiagnosed despite increasing public awareness. The condition is characterized by insulin resistance, the development of diabetes at a young age (before 25 years), mild high blood sugar levels, inheritance in an autosomal dominant pattern, and the preservation of natural insulin production. RESULTS Currently, there are 14 distinct subtypes of MODY that have been identified. Each subtype possesses distinct characteristics in terms of their frequency, clinical symptoms, severity of diabetes, related complications, and response to medicinal interventions. Due to the clinical similarities, lack of awareness, and high expense of genetic testing, distinguishing between type I (T1D) and type II diabetes mellitus (T2D) can be challenging, resulting in misdiagnosis of this type of diabetes. As a consequence, a significant number of individuals are being deprived of adequate medical attention. Accurate diagnosis enables the utilization of novel therapeutic strategies and enhances the management of therapy in comparison to type II and type I diabetes. CONCLUSION This article offers a concise overview of the clinical subtypes and characteristics of monogenic diabetes. Furthermore, this article discusses the various subtypes of MODY, as well as the process of diagnosing, managing, and treating the condition. It also addresses the difficulties encountered in detecting and treating MODY.
Collapse
Affiliation(s)
- Manisha Sharma
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Kajal Maurya
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | - Anuj Nautiyal
- Department of Pharmacy Practice, School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Dehradun, Uttarakhand, India
| | | |
Collapse
|
4
|
Luckett AM, Hawkes G, Green HD, De Franco E, Hagopian WA, Roep BO, Weedon MN, Oram RA, Johnson MB. Type 1 Diabetes Genetic Risk Contributes to Phenotypic Presentation in Monogenic Autoimmune Diabetes. Diabetes 2025; 74:243-248. [PMID: 39531505 PMCID: PMC11755682 DOI: 10.2337/db24-0485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
ARTICLE HIGHLIGHTS There is variability in early-onset autoimmune diabetes presentation in individuals with monogenic autoimmunity; the mechanism(s) underlying this is unclear. We examined whether type 1 diabetes (T1D) polygenic risk contributes to clinical phenotype in monogenic autoimmune diabetes. Individuals with monogenic autoimmune diabetes had higher T1D genetic risk scores compared with control cohorts, driven largely by increased presence of T1D-risk DR3-DQ2 haplotype. Established T1D polygenic risk alleles, particularly class II HLA genes, contribute to clinical presentation in monogenic autoimmunity.
Collapse
Affiliation(s)
- Amber M. Luckett
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
| | - Gareth Hawkes
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
| | - Harry D. Green
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
| | - Elisa De Franco
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
| | - William A. Hagopian
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Bart O. Roep
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Michael N. Weedon
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
| | - Richard A. Oram
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
- Royal Devon University Healthcare NHS Foundation Trust, Exeter, U.K
| | - Matthew B. Johnson
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, U.K
| |
Collapse
|
5
|
Hughes J, Shymka M, Ng T, Phulka JS, Safabakhsh S, Laksman Z. Polygenic Risk Score Implementation into Clinical Practice for Primary Prevention of Cardiometabolic Disease. Genes (Basel) 2024; 15:1581. [PMID: 39766848 PMCID: PMC11675431 DOI: 10.3390/genes15121581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Cardiovascular disease is a leading cause of mortality globally and a major contributor to disability. Traditional risk factors, as initially established in the FRAMINGHAM study, have helped to stratify populations and identify patients for early intervention. Incorporating genetic factors enhances risk stratification tools, enabling the earlier identification of individuals at increased risk and facilitating more targeted and effective risk factor modifications. While monogenic risk variants are present in a minority of the population, polygenic risk scores (PRS) are collections of multiple single-nucleotide variants that collectively provide summative risk and capture a more accurate risk score for a greater number of people. PRS have demonstrated clear utility in cardiometabolic diseases by predicting onset, progression, and therapeutic response. Methods: A structured and exploratory hybrid search strategy was employed, combining keyword-based database searches and supplementary techniques to comprehensively synthesize the literature on PRS implementation in clinical practice. Discussion: A comprehensive overview of PRS in cardiometabolic diseases and their potential avenues for integration into primary care is discussed. First, we examine the implementation of genetic screening, risk communication, and intervention strategies through the lens of the American Heart Association's implementation criteria, focusing on their efficacy, minimization of harm, and logistical considerations. Then, we explores how the varied perceptions of patients and practitioners towards PRS can influence both adoption and utilization. Lastly, we addresses the need for the development of clear guidelines and regulations to support this process, ensuring PRS integration is both scientifically sound and ethically responsible. Future directions: Initiatives aimed at advancing personalized approaches to disease prevention will enhance health outcomes. Developing guidelines for the responsible use of PRS by establishing benefits, while mitigating risk, will a key factor in implementation for clinical utility. Conclusions: For integration into clinical practice, we must address both patient and provider concerns and experience. Standardized guidelines and training will help to effectively implement PRS into clinical practice. Developing these resources will be essential for PRS to fulfill its potential in personalized, patient-centered care.
Collapse
Affiliation(s)
- Julia Hughes
- Department of Medicine, University of British Columbia, Vancouver, BC V5M 1M9, Canada; (J.H.); (M.S.); (T.N.)
| | - Mikayla Shymka
- Department of Medicine, University of British Columbia, Vancouver, BC V5M 1M9, Canada; (J.H.); (M.S.); (T.N.)
| | - Trevor Ng
- Department of Medicine, University of British Columbia, Vancouver, BC V5M 1M9, Canada; (J.H.); (M.S.); (T.N.)
| | - Jobanjit S. Phulka
- Department of Medicine, University of British Columbia, Vancouver, BC V5M 1M9, Canada; (J.H.); (M.S.); (T.N.)
| | - Sina Safabakhsh
- Department of Medicine, University of British Columbia, Vancouver, BC V5M 1M9, Canada; (J.H.); (M.S.); (T.N.)
| | - Zachary Laksman
- Department of Medicine, University of British Columbia, Vancouver, BC V5M 1M9, Canada; (J.H.); (M.S.); (T.N.)
- Centre for Heart Lung Innovation (HLI), Vancouver, BC V6Z 1Y6, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| |
Collapse
|
6
|
Ribeiro AF, Fitas AL, Pires MO, Matoso P, Ligeiro D, Sobral D, Penha-Gonçalves C, Demengeot J, Caramalho Í, Limbert C. Whole Exome Sequencing in Children With Type 1 Diabetes Before Age 6 Years Reveals Insights Into Disease Heterogeneity. J Diabetes Res 2024; 2024:3076895. [PMID: 39364395 PMCID: PMC11449554 DOI: 10.1155/2024/3076895] [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: 02/28/2024] [Revised: 06/04/2024] [Accepted: 08/24/2024] [Indexed: 10/05/2024] Open
Abstract
Aims: This study is aimed at comparing whole exome sequencing (WES) data with the clinical presentation in children with type 1 diabetes onset ≤ 5 years of age (EOT1D). Methods: WES was performed in 99 unrelated children with EOT1D with subsequent analysis to identify potentially deleterious rare variants in MODY genes. High-resolution HLA class II haplotyping, SNP genotyping, and T1D-genetic risk score (T1D-GRS) were also evaluated. Results: Eight of the ninety-nine EOT1D participants carried a potentially deleterious rare variant in a MODY gene. Rare variants affected five genes: GCK (n = 1), HNF1B (n = 2), HNF4A (n = 1), PDX1 (n = 2), and RFX6 (n = 2). At diagnosis, these children had a mean age of 3.0 years, a mean HbA1c of 10.5%, a detectable C-peptide in 5/8, and a positive islet autoantibody in 6/7. Children with MODY variants tend to exhibit a lower number of pancreatic autoantibodies and a lower fasting C-peptide compared to EOT1D without MODY rare variants. They also carried at least one high-risk DR3-DQ2 or DR4-DQ8 haplotype and exhibited a T1D-GRS similar to the other individuals in the EOT1D cohort, but higher than healthy controls. Conclusions: WES found potentially deleterious rare variants in MODY genes in 8.1% of EOT1D, occurring in the context of a T1D genetic background. Such genetic variants may contribute to disease precipitation by a β-cell dysfunction mechanism. This supports the concept of different endotypes of T1D, and WES at T1D onset may be a prerequisite for the implementation of precision therapies in children with autoimmune diabetes.
Collapse
Affiliation(s)
- Andreia Fiúza Ribeiro
- Pediatric Endocrinology UnitHospital de Dona EstefâniaSão José Local Health Unit, Lisbon, Portugal
- Pediatric DepartmentHospital Prof. Doutor Fernando FonsecaAmadora Sintra Local Health Unit, Amadora, Portugal
| | - Ana Laura Fitas
- Pediatric Endocrinology UnitHospital de Dona EstefâniaSão José Local Health Unit, Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC)NOVA Medical SchoolUniversidade NOVA de Lisboa, Lisbon, Portugal
| | - Marcela Oliveira Pires
- Pediatric Endocrinology UnitHospital de Dona EstefâniaSão José Local Health Unit, Lisbon, Portugal
- Pediatric DepartmentHospital de São Francisco XavierLisboa Ocidental Local Health Unit, Lisbon, Portugal
| | - Paula Matoso
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Dário Ligeiro
- Blood and Transplantation Center of LisbonInstituto Português do Sangue e da Transplantação, Lisbon, Portugal
- Immunosurgery UnitChampalimaud Foundation, Lisbon, Portugal
| | | | | | | | - Íris Caramalho
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- Faculty of SciencesUniversity of Lisbon, Lisbon, Portugal
| | - Catarina Limbert
- Pediatric Endocrinology UnitHospital de Dona EstefâniaSão José Local Health Unit, Lisbon, Portugal
- Comprehensive Health Research Centre (CHRC)NOVA Medical SchoolUniversidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
7
|
Noble JA. Fifty years of HLA-associated type 1 diabetes risk: history, current knowledge, and future directions. Front Immunol 2024; 15:1457213. [PMID: 39328411 PMCID: PMC11424550 DOI: 10.3389/fimmu.2024.1457213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 08/16/2024] [Indexed: 09/28/2024] Open
Abstract
More than 50 years have elapsed since the association of human leukocyte antigens (HLA) with type 1 diabetes (T1D) was first reported. Since then, methods for identification of HLA have progressed from cell based to DNA based, and the number of recognized HLA variants has grown from a few to tens of thousands. Current genotyping methodology allows for exact identification of all HLA-encoding genes in an individual's genome, with statistical analysis methods evolving to digest the enormous amount of data that can be produced at an astonishing rate. The HLA region of the genome has been repeatedly shown to be the most important genetic risk factor for T1D, and the original reported associations have been replicated, refined, and expanded. Even with the remarkable progress through 50 years and over 5,000 reports, a comprehensive understanding of all effects of HLA on T1D remains elusive. This report represents a summary of the field as it evolved and as it stands now, enumerating many past and present challenges, and suggests possible paradigm shifts for moving forward with future studies in hopes of finally understanding all the ways in which HLA influences the pathophysiology of T1D.
Collapse
Affiliation(s)
- Janelle A. Noble
- Children’s Hospital Oakland Research Institute,
Oakland, CA, United States
- University of California San Francisco, Oakland,
CA, United States
| |
Collapse
|
8
|
Szczerbinski L, Florez JC. Precision medicine in diabetes - current trends and future directions. Is the future now? COMPREHENSIVE PRECISION MEDICINE 2024:458-483. [DOI: 10.1016/b978-0-12-824010-6.00021-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Venkatachari IV, Chougule A, Gowri V, Taur P, Bodhanwala M, Prabhu S, Madkaikar M, Desai M. Monogenic inborn errors of immunity in autoimmune disorders. Immunol Res 2023; 71:771-780. [PMID: 37199901 DOI: 10.1007/s12026-023-09391-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/03/2023] [Indexed: 05/19/2023]
Abstract
To estimate the prevalence of monogenic inborn errors of immunity in patients with autoimmune diseases (AID), the study included 56 subjects (male:female ratio: 1.07) with mean age of onset of autoimmunity 7 years (4 months-46 years). 21/56 had polyautoimmunity. 5/56 patients met the JMF criteria for PID. The different AID referred were hematological (42%) > gastrointestinal (GI) (16%) > skin (14%) > endocrine (10%) > rheumatological (8%) > renal (6%) > neurological (2%). 36/56 reported recurrent infections. 27/56 were on polyimmunotherapy. 18/52 (35%) had CD19 lymphopenia, 24/52 (46%) had CD4 lymphopenia, 11/52 (21%) had CD8 lymphopenia, and 14/48 (29%) had NK lymphopenia. 21/50 (42%) had hypogammaglobinemia; 3 of whom were given rituximab. 28/56 were found to have pathogenic variants among PIRD genes. These 28 patients had 42 AID among which hematological was most common (50%) > GI (14%) = skin (14%)> endocrine (9%) > rheumatological (7%) > renal and neurological (2%). Hematological AID was the most common AID (75%) in children with PIRD. Positive predictive value (PPV) of abnormal immunological tests was 50% and sensitivity of 70%. JMF criteria had specificity of 100% in identifying PIRD and sensitivity of 17%. Polyautoimmunity had a PPV of 35% and sensitivity of 40%. 11/28 of these children were offered transplant. 8/28 were started on sirolimus, 2/28 on abatacept, and 3/28 on baricitinib/ruxolitinib after diagnosis. In conclusion, 50% of children with AID have underlying PIRD. LRBA deficiency and STAT1 GOF were the most common PIRD. Age at presentation, number of autoimmunity, routine immunological tests, and JMF criteria are not predictive of underlying PIRD. Early diagnosis with exome sequencing alters the prognosis and opens new therapeutic avenue.
Collapse
Affiliation(s)
| | - Akshaya Chougule
- Department of Immunology, Bai Jerbai Wadia Hospital for Children, Acharya Dhonde Marg, Parel, Mumbai, 400012, India
| | - Vijaya Gowri
- Department of Immunology, Bai Jerbai Wadia Hospital for Children, Acharya Dhonde Marg, Parel, Mumbai, 400012, India
| | - Prasad Taur
- Department of Immunology, Bai Jerbai Wadia Hospital for Children, Acharya Dhonde Marg, Parel, Mumbai, 400012, India
| | - Minnie Bodhanwala
- Department of Immunology, Bai Jerbai Wadia Hospital for Children, Acharya Dhonde Marg, Parel, Mumbai, 400012, India
| | - Shakuntala Prabhu
- Department of Immunology, Bai Jerbai Wadia Hospital for Children, Acharya Dhonde Marg, Parel, Mumbai, 400012, India
| | - Manisha Madkaikar
- National Institute of Immunohematology, ICMR, KEM, Parel, Mumbai, India
| | - Mukesh Desai
- Department of Immunology, Bai Jerbai Wadia Hospital for Children, Acharya Dhonde Marg, Parel, Mumbai, 400012, India.
| |
Collapse
|
10
|
Mancuso G, Bechi Genzano C, Fierabracci A, Fousteri G. Type 1 diabetes and inborn errors of immunity: Complete strangers or 2 sides of the same coin? J Allergy Clin Immunol 2023; 151:1429-1447. [PMID: 37097271 DOI: 10.1016/j.jaci.2023.03.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/26/2023]
Abstract
Type 1 diabetes (T1D) is a polygenic disease and does not follow a mendelian pattern. Inborn errors of immunity (IEIs), on the other hand, are caused by damaging germline variants, suggesting that T1D and IEIs have nothing in common. Some IEIs, resulting from mutations in genes regulating regulatory T-cell homeostasis, are associated with elevated incidence of T1D. The genetic spectrum of IEIs is gradually being unraveled; consequently, molecular pathways underlying human monogenic autoimmunity are being identified. There is an appreciable overlap between some of these pathways and the genetic variants that determine T1D susceptibility, suggesting that after all, IEI and T1D are 2 sides of the same coin. The study of monogenic IEIs with a variable incidence of T1D has the potential to provide crucial insights into the mechanisms leading to T1D. These insights contribute to the definition of T1D endotypes and explain disease heterogeneity. In this review, we discuss the interconnected pathogenic pathways of autoimmunity, β-cell function, and primary immunodeficiency. We also examine the role of environmental factors in disease penetrance as well as the circumstantial evidence of IEI drugs in preventing and curing T1D in individuals with IEIs, suggesting the repositioning of these drugs also for T1D therapy.
Collapse
Affiliation(s)
- Gaia Mancuso
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Camillo Bechi Genzano
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | | | - Georgia Fousteri
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
11
|
Gray PE, David C. Inborn Errors of Immunity and Autoimmune Disease. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:1602-1622. [PMID: 37119983 DOI: 10.1016/j.jaip.2023.04.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/01/2023] [Accepted: 04/21/2023] [Indexed: 05/01/2023]
Abstract
Autoimmunity may be a manifestation of inborn errors of immunity, specifically as part of the subgroup of primary immunodeficiency known as primary immune regulatory disorders. However, although making a single gene diagnosis can have important implications for prognosis and management, picking patients to screen can be difficult, against a background of a high prevalence of autoimmune disease in the population. This review compares the genetics of common polygenic and rare monogenic autoimmunity, and explores the molecular mechanisms, phenotypes, and inheritance of autoimmunity associated with primary immune regulatory disorders, highlighting the emerging importance of gain-of-function and non-germline somatic mutations. A novel framework for identifying rare monogenic cases of common diseases in children is presented, highlighting important clinical and immunologic features that favor single gene disease and guides clinicians in selecting appropriate patients for genomic screening. In addition, there will be a review of autoimmunity in non-genetically defined primary immunodeficiency such as common variable immunodeficiency, and of instances where primary autoimmunity can result in clinical phenocopies of inborn errors of immunity.
Collapse
Affiliation(s)
- Paul Edgar Gray
- Sydney Children's Hospital, Randwick, NSW, Australia; Western Sydney University, Penrith, NSW, Australia.
| | - Clementine David
- Sydney Children's Hospital, Randwick, NSW, Australia; The School of Women's & Children's Health, University of New South Wales, Randwick, NSW, Australia
| |
Collapse
|
12
|
Peters LD, Yeh WI, Arnoletti JM, Brown ME, Posgai AL, Mathews CE, Brusko TM. Modeling cell-mediated immunity in human type 1 diabetes by engineering autoreactive CD8 + T cells. Front Immunol 2023; 14:1142648. [PMID: 37325626 PMCID: PMC10262917 DOI: 10.3389/fimmu.2023.1142648] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/13/2023] [Indexed: 06/17/2023] Open
Abstract
The autoimmune pathogenesis of type 1 diabetes (T1D) involves cellular infiltration from innate and adaptive immune subsets into the islets of Langerhans within the pancreas; however, the direct cytotoxic killing of insulin-producing β-cells is thought to be mediated primarily by antigen-specific CD8+ T cells. Despite this direct pathogenic role, key aspects of their receptor specificity and function remain uncharacterized, in part, due to their low precursor frequency in peripheral blood. The concept of engineering human T cell specificity, using T cell receptor (TCR) and chimeric antigen receptor (CAR)-based approaches, has been demonstrated to improve adoptive cell therapies for cancer, but has yet to be extensively employed for modeling and treating autoimmunity. To address this limitation, we sought to combine targeted genome editing of the endogenous TCRα chain gene (TRAC) via CRISPR/Cas9 in combination with lentiviral vector (LV)-mediated TCR gene transfer into primary human CD8+ T cells. We observed that knockout (KO) of endogenous TRAC enhanced de novo TCR pairing, which permitted increased peptide:MHC-dextramer staining. Moreover, TRAC KO and TCR gene transfer increased markers of activation and effector function following activation, including granzyme B and interferon-γ production. Importantly, we observed increased cytotoxicity toward an HLA-A*0201+ human β-cell line by HLA-A*02:01 restricted CD8+ T cells engineered to recognize islet-specific glucose-6-phosphatase catalytic subunit (IGRP). These data support the notion of altering the specificity of primary human T cells for mechanistic analyses of autoreactive antigen-specific CD8+ T cells and are expected to facilitate downstream cellular therapeutics to achieve tolerance induction through the generation of antigen-specific regulatory T cells.
Collapse
Affiliation(s)
- Leeana D. Peters
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Wen-I Yeh
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Juan M. Arnoletti
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Matthew E. Brown
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Amanda L. Posgai
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Clayton E. Mathews
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Todd M. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
- Department of Pediatrics, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
13
|
Carrera P, Marzinotto I, Bonfanti R, Massimino L, Calzavara S, Favellato Μ, Jofra T, De Giglio V, Bonura C, Stabilini A, Favalli V, Bondesan S, Cicalese MP, Laurenzi A, Caretto A, Frontino G, Rigamonti A, Molinari C, Scavini M, Sandullo F, Zapparoli E, Caridi N, Bonfiglio S, Castorani V, Ungaro F, Petrelli A, Barera G, Aiuti A, Bosi E, Battaglia M, Piemonti L, Lampasona V, Fousteri G. Genetic determinants of type 1 diabetes in individuals with weak evidence of islet autoimmunity at disease onset. Diabetologia 2023; 66:695-708. [PMID: 36692510 DOI: 10.1007/s00125-022-05865-5] [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: 03/18/2022] [Accepted: 10/31/2022] [Indexed: 01/25/2023]
Abstract
AIMS/HYPOTHESIS Islet autoantibodies (AAbs) are detected in >90% of individuals with clinically suspected type 1 diabetes at disease onset. A single AAb, sometimes at low titre, is often detected in some individuals, making their diagnosis uncertain. Type 1 diabetes genetic risk scores (GRS) are a useful tool for discriminating polygenic autoimmune type 1 diabetes from other types of diabetes, particularly the monogenic forms, but testing is not routinely performed in the clinic. Here, we used a type 1 diabetes GRS to screen for monogenic diabetes in individuals with weak evidence of autoimmunity, i.e. with a single AAb at disease onset. METHODS In a pilot study, we genetically screened 142 individuals with suspected type 1 diabetes, 42 of whom were AAb-negative, 27 of whom had a single AAb (single AAb-positive) and 73 of whom had multiple AAbs (multiple AAb-positive) at disease onset. Next-generation sequencing (NGS) was performed in 41 AAb-negative participants, 26 single AAb-positive participants and 60 multiple AAb-positive participants using an analysis pipeline of more than 200 diabetes-associated genes. RESULTS The type 1 diabetes GRS was significantly lower in AAb-negative individuals than in those with a single and multiple AAbs. Pathogenetic class 4/5 variants in MODY or monogenic diabetes genes were identified in 15/41 (36.6%) AAb-negative individuals, while class 3 variants of unknown significance were identified in 17/41 (41.5%). Residual C-peptide levels at diagnosis were higher in individuals with mutations compared to those without pathogenetic variants. Class 3 variants of unknown significance were found in 11/26 (42.3%) single AAb-positive individuals, and pathogenetic class 4/5 variants were present in 2/26 (7.7%) single AAb-positive individuals. No pathogenetic class 4/5 variants were identified in multiple AAb-positive individuals, but class 3 variants of unknown significance were identified in 19/60 (31.7%) patients. Several patients across the three groups had more than one class 3 variant. CONCLUSIONS/INTERPRETATION These findings provide insights into the genetic makeup of patients who show weak evidence of autoimmunity at disease onset. Absence of islet AAbs or the presence of a single AAb together with a low type 1 diabetes GRS may be indicative of a monogenic form of diabetes, and use of NGS may improve the accuracy of diagnosis.
Collapse
Affiliation(s)
- Paola Carrera
- Unit of Genomics for Human Disease Diagnosis, IRCCS Ospedale San Raffaele, Milan, Italy
- Laboratory of Clinical Molecular Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ilaria Marzinotto
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Riccardo Bonfanti
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Luca Massimino
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele Hospital, Milan, Italy
| | - Silvia Calzavara
- Laboratory of Clinical Molecular Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Tatiana Jofra
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Clara Bonura
- Pediatric Department, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Angela Stabilini
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Valeria Favalli
- Pediatric Department, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Simone Bondesan
- Unit of Genomics for Human Disease Diagnosis, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Maria Pia Cicalese
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Laurenzi
- Department of Internal Medicine, Diabetology, Endocrinology and Metabolism, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Amelia Caretto
- Department of Internal Medicine, Diabetology, Endocrinology and Metabolism, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giulio Frontino
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Andrea Rigamonti
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Chiara Molinari
- Department of Internal Medicine, Diabetology, Endocrinology and Metabolism, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Marina Scavini
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Department of Internal Medicine, Diabetology, Endocrinology and Metabolism, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Federica Sandullo
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ettore Zapparoli
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicoletta Caridi
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Bonfiglio
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Federica Ungaro
- Department of Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele Hospital, Milan, Italy
| | | | - Graziano Barera
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Pediatric Department, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessandro Aiuti
- Vita-Salute San Raffaele University, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Emanuele Bosi
- Department of Internal Medicine, Diabetology, Endocrinology and Metabolism, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Manuela Battaglia
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Fondazione Telethon, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Vito Lampasona
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy.
| | - Georgia Fousteri
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
14
|
Narula M, Lakshmanan U, Borna S, Schulze JJ, Holmes TH, Harre N, Kirkey M, Ramachandran A, Tagi VM, Barzaghi F, Grunebaum E, Upton JEM, Hong-Diep Kim V, Wysocki C, Dimitriades VR, Weinberg K, Weinacht KG, Gernez Y, Sathi BK, Schelotto M, Johnson M, Olek S, Sachsenmaier C, Roncarolo MG, Bacchetta R. Epigenetic and immunological indicators of IPEX disease in subjects with FOXP3 gene mutation. J Allergy Clin Immunol 2023; 151:233-246.e10. [PMID: 36152823 DOI: 10.1016/j.jaci.2022.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/16/2022] [Accepted: 09/09/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Forkhead box protein 3 (FOXP3) is the master transcription factor in CD4+CD25hiCD127lo regulatory T (Treg) cells. Mutations in FOXP3 result in IPEX (immune dysregulation, polyendocrinopathy, enteropathy, X-linked) syndrome. Clinical presentation of IPEX syndrome is broader than initially described, challenging the understanding of the disease, its evolution, and treatment choice. OBJECTIVE We sought to study the type and extent of immunologic abnormalities that remain ill-defined in IPEX, across genetic and clinical heterogeneity. METHODS We performed Treg-cell-specific epigenetic quantification and immunologic characterization of severe "typical" (n = 6) and "atypical" or asymptomatic (n = 9) patients with IPEX. RESULTS Increased number of cells with Treg-cell-Specific Demethylated Region demethylation in FOXP3 is a consistent feature in patients with IPEX, with (1) highest values in those with typical IPEX, (2) increased values in subjects with pathogenic FOXP3 but still no symptoms, and (3) gradual increase over the course of disease progression. Large-scale profiling using Luminex identified plasma inflammatory signature of macrophage activation and TH2 polarization, with cytokines previously not associated with IPEX pathology, including CCL22, CCL17, CCL15, and IL-13, and the inflammatory markers TNF-α, IL-1A, IL-8, sFasL, and CXCL9. Similarly, both Treg-cell and Teff compartments, studied by Mass Cytometry by Time-Of-Flight, were skewed toward the TH2 compartment, especially in typical IPEX. CONCLUSIONS Elevated TSDR-demethylated cells, combined with elevation of plasmatic and cellular markers of a polarized type 2 inflammatory immune response, extends our understanding of IPEX diagnosis and heterogeneity.
Collapse
Affiliation(s)
- Mansi Narula
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Uma Lakshmanan
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Simon Borna
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, Calif
| | | | - Tyson H Holmes
- Human Immune Monitoring Center, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, Calif
| | - Nicholas Harre
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Matthew Kirkey
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Akshaya Ramachandran
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Veronica Maria Tagi
- San Raffaele Telethon Institute for Gene Therapy, Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute Milan, Milan, Italy
| | - Federica Barzaghi
- San Raffaele Telethon Institute for Gene Therapy, Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute Milan, Milan, Italy
| | - Eyal Grunebaum
- Division of Immunology and Allergy, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Julia E M Upton
- Division of Immunology and Allergy, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Vy Hong-Diep Kim
- Division of Immunology and Allergy, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Christian Wysocki
- Department of Internal Medicine, Pediatrics, Allergy and Immunology, UT Southwestern Medical Center, Dallas, Tex
| | - Victoria R Dimitriades
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, UC Davis Health Medical Center, Sacramento, Calif
| | - Kenneth Weinberg
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Katja G Weinacht
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Yael Gernez
- Department of Pediatrics, Division of Allergy, Immunology, and Rheumatology, Stanford University School of Medicine, Stanford, Calif
| | | | | | - Matthew Johnson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, United Kingdom
| | - Sven Olek
- Ivana Turbachova Laboratory of Epigenetics, Precision for Medicine GmbH, Berlin, Germany
| | | | - Maria-Grazia Roncarolo
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, Calif; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, Calif; Center for Definitive and Curative Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Rosa Bacchetta
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Stanford University School of Medicine, Stanford, Calif; Center for Definitive and Curative Medicine, Stanford University School of Medicine, Stanford, Calif.
| |
Collapse
|
15
|
Abstract
The historical subclassification of diabetes into predominantly types 1 and 2 is well appreciated to inadequately capture the heterogeneity seen in patient presentations, disease course, response to therapy and disease complications. This review summarises proposed data-driven approaches to further refine diabetes subtypes using clinical phenotypes and/or genetic information. We highlight the benefits as well as the limitations of these subclassification schemas, including practical barriers to their implementation that would need to be overcome before incorporation into clinical practice.
Collapse
Affiliation(s)
- Aaron J Deutsch
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical & Population Genetics, Broad Institute, Boston, MA, USA
- Program in Metabolism, Broad Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Emma Ahlqvist
- Genomics, Diabetes and Endocrinology, Department of Clinical Sciences in Malmö, Lund University Diabetes Centre, Lund University, Malmö, Sweden.
| | - Miriam S Udler
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Program in Medical & Population Genetics, Broad Institute, Boston, MA, USA.
- Program in Metabolism, Broad Institute, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Di Lorenzo B, Pacillo L, Milardi G, Jofra T, Di Cesare S, Gerosa J, Marzinotto I, Zapparoli E, Rivalta B, Cifaldi C, Barzaghi F, Giancotta C, Zangari P, Rapini N, Deodati A, Amodio G, Passerini L, Carrera P, Gregori S, Palma P, Finocchi A, Lampasona V, Cicalese MP, Schiaffini R, Di Matteo G, Merelli I, Barcella M, Aiuti A, Piemonti L, Cancrini C, Fousteri G. Natural history of type 1 diabetes on an immunodysregulatory background with genetic alteration in B-cell activating factor receptor: A case report. Front Immunol 2022; 13:952715. [PMID: 36090979 PMCID: PMC9459137 DOI: 10.3389/fimmu.2022.952715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022] Open
Abstract
The immunological events leading to type 1 diabetes (T1D) are complex and heterogeneous, underscoring the necessity to study rare cases to improve our understanding. Here, we report the case of a 16-year-old patient who showed glycosuria during a regular checkup. Upon further evaluation, stage 2 T1D, autoimmune thrombocytopenic purpura (AITP), and common variable immunodeficiency (CVID) were diagnosed. The patient underwent low carb diet, losing > 8 kg, and was placed on Ig replacement therapy. Anti-CD20 monoclonal antibody (Rituximab, RTX) was administered 2 years after diagnosis to treat peripheral polyneuropathy, whereas an atypical mycobacteriosis manifested 4 years after diagnosis and was managed with prolonged antibiotic treatment. In the fifth year of monitoring, the patient progressed to insulin dependency despite ZnT8A autoantibody resolution and IA-2A and GADA autoantibody decline. The patient had low T1D genetic risk score (GRS = 0.22817) and absence of human leukocyte antigen (HLA) DR3/DR4-DQ8. Genetic analysis identified the monoallelic mutation H159Y in TNFRSF13C, a gene encoding B-cell activating factor receptor (BAFFR). Significant reduced blood B-cell numbers and BAFFR levels were observed in line with a dysregulation in BAFF–BAFFR signaling. The elevated frequency of PD-1+ dysfunctional Tfh cells composed predominantly by Th1 phenotype was observed at disease onset and during follow-up. This case report describes a patient progressing to T1D on a BAFFR-mediated immunodysregulatory background, suggesting a role of BAFF–BAFFR signaling in islet-specific tolerance and T1D progression.
Collapse
Affiliation(s)
- Biagio Di Lorenzo
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele, Milan, Italy
| | - Lucia Pacillo
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesú Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Giulia Milardi
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele, Milan, Italy
| | - Tatiana Jofra
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele, Milan, Italy
| | - Silvia Di Cesare
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Jolanda Gerosa
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele, Milan, Italy
| | - Ilaria Marzinotto
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele, Milan, Italy
| | - Ettore Zapparoli
- Center for Omics Sciences, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele, Milano, Italy
| | - Beatrice Rivalta
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesú Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Cristina Cifaldi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesú Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Federica Barzaghi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Carmela Giancotta
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesú Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Paola Zangari
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesú Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Novella Rapini
- Unit of Endocrinology, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Annalisa Deodati
- Unit of Endocrinology, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Giada Amodio
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Laura Passerini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Paola Carrera
- Unit of Genomics for Human Disease Diagnosis and Laboratory of Clinical Molecular Biology, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele, Milan, Italy
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Palma
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesú Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Andrea Finocchi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesú Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Vito Lampasona
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele, Milan, Italy
| | - Maria Pia Cicalese
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Faculty of Medicine, University Vita-Salute San Raffaele, Milan, Italy
| | - Riccardo Schiaffini
- Unit of Endocrinology, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Gigliola Di Matteo
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Department of Bioinformatics, Institute for Biomedical Technologies National Research Council, Segrate, Italy
| | - Matteo Barcella
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Department of Bioinformatics, Institute for Biomedical Technologies National Research Council, Segrate, Italy
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- Faculty of Medicine, University Vita-Salute San Raffaele, Milan, Italy
- *Correspondence: Alessandro Aiuti, ; Caterina Cancrini, ; Georgia Fousteri, ; Lorenzo Piemonti,
| | - Lorenzo Piemonti
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele, Milan, Italy
- *Correspondence: Alessandro Aiuti, ; Caterina Cancrini, ; Georgia Fousteri, ; Lorenzo Piemonti,
| | - Caterina Cancrini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesú Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
- *Correspondence: Alessandro Aiuti, ; Caterina Cancrini, ; Georgia Fousteri, ; Lorenzo Piemonti,
| | - Georgia Fousteri
- Diabetes Research Institute, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele, Milan, Italy
- *Correspondence: Alessandro Aiuti, ; Caterina Cancrini, ; Georgia Fousteri, ; Lorenzo Piemonti,
| |
Collapse
|
17
|
Colclough K, van Heugten R, Patel K. An update on the diagnosis and management of monogenic diabetes. PRACTICAL DIABETES 2022. [DOI: 10.1002/pdi.2410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Kevin Colclough
- Exeter Genomics Laboratory, Royal Devon & Exeter NHS Foundation Trust Exeter UK
| | - Rachel van Heugten
- Exeter Genomics Laboratory, Royal Devon & Exeter NHS Foundation Trust Exeter UK
| | - Kashyap Patel
- Institute of Biomedical and Clinical Science University of Exeter Medical School Exeter UK
| |
Collapse
|
18
|
Wyatt RC, Hagopian WA, Roep BO, Patel KA, Resnick B, Dobbs R, Hudson M, De Franco E, Ellard S, Flanagan SE, Hattersley AT, Oram RA, Johnson MB. Congenital beta cell defects are not associated with markers of islet autoimmunity, even in the context of high genetic risk for type 1 diabetes. Diabetologia 2022; 65:1179-1184. [PMID: 35501400 PMCID: PMC9174109 DOI: 10.1007/s00125-022-05697-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 02/14/2022] [Indexed: 11/14/2022]
Abstract
AIMS/HYPOTHESIS A key unanswered question in type 1 diabetes is whether beta cells initiate their own destruction or are victims of an aberrant immune response (beta cell suicide or homicide?). To investigate this, we assessed islet autoantibodies in individuals with congenital beta cell defects causing neonatal diabetes mellitus (NDM). METHODS We measured autoantibodies to GAD (GADA), islet antigen-2 (IA-2A) and zinc transporter 8 (ZnT8A) in 242 individuals with NDM (median age diagnosed 1.8 months [IQR 0.39-2.9 months]; median age collected 4.6 months [IQR 1.8-27.6 months]; median diabetes duration 2 months [IQR 0.6-23 months]), including 75 whose NDM resulted from severe beta cell endoplasmic reticulum (ER) stress. As a control cohort we also tested samples from 69 diabetes-free individuals (median age collected 9.9 months [IQR 9.0-48.6 months]) for autoantibodies. RESULTS We found low prevalence of islet autoantibodies in individuals with monogenic NDM; 13/242 (5.4% [95% CI 2.9, 9.0%]) had detectable GADA, IA-2A and/or ZnT8A. This was similar to the proportion in the control participants who did not have diabetes (1/69 positive [1.4%, 95% CI 0.03, 7.8%], p=0.3). Importantly, monogenic individuals with beta cell ER stress had a similar rate of GADA/IA-2A/ZnT8A positivity to non-ER stress aetiologies (2.7% [95% CI 0.3, 9.3%] vs 6.6% [95% CI 3.3, 11.5%] p=0.4). We observed no association between islet autoimmunity and genetic risk, age at testing (including 30 individuals >10 years at testing) or diabetes duration (p>0.4 for all). CONCLUSIONS/INTERPRETATION Our data support the hypothesis that beta cell stress/dysfunction alone does not lead to the production of islet autoantibodies, even in the context of high-risk HLA types. This suggests that additional factors are required to trigger an autoimmune response towards beta cells.
Collapse
Affiliation(s)
- Rebecca C Wyatt
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | | | - Bart O Roep
- Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Kashyap A Patel
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Brittany Resnick
- National Institute for Health Research Exeter Clinical Research Facility, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Rebecca Dobbs
- National Institute for Health Research Exeter Clinical Research Facility, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Michelle Hudson
- National Institute for Health Research Exeter Clinical Research Facility, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Elisa De Franco
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Sian Ellard
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Sarah E Flanagan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Andrew T Hattersley
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Richard A Oram
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Matthew B Johnson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
19
|
Basmanav FB, Betz RC. Translational impact of omics studies in alopecia areata: recent advances and future perspectives. Expert Rev Clin Immunol 2022; 18:845-857. [PMID: 35770930 DOI: 10.1080/1744666x.2022.2096590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Alopecia areata (AA) is a non-scarring, hair loss disorder and a common autoimmune-mediated disease with an estimated lifetime risk of about 2%. To date, the treatment of AA is mainly based on suppression or stimulation of the immune response. Genomics and transcriptomics studies generated important insights into the underlying pathophysiology, enabled discovery of molecular disease signatures, which were used in some of the recent clinical trials to monitor drug response and substantiated the consideration of new therapeutic modalities for the treatment of AA such as abatacept, dupilumab, ustekinumab and Janus Kinase (JAK) inhibitors. AREAS COVERED In this review, genomics and transcriptomics studies in AA are discussed in detail with particular emphasis on their past and prospective translational impacts. Microbiome studies are also briefly introduced. EXPERT OPINION The generation of large datasets using the new high-throughput technologies has revolutionized medical research and AA has also benefited from the wave of omics studies. However, the limitations associated with JAK inhibitors and clinical heterogeneity in AA patients underscore the necessity for continuing omics research in AA for discovery of novel therapeutic modalities and development of clinical tools for precision medicine.
Collapse
Affiliation(s)
- F Buket Basmanav
- Medical Faculty & University Hospital Bonn, Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Regina C Betz
- Medical Faculty & University Hospital Bonn, Institute of Human Genetics, University of Bonn, Bonn, Germany
| |
Collapse
|
20
|
Barroso I. The importance of increasing population diversity in genetic studies of type 2 diabetes and related glycaemic traits. Diabetologia 2021; 64:2653-2664. [PMID: 34595549 PMCID: PMC8563561 DOI: 10.1007/s00125-021-05575-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 07/07/2021] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes has a global prevalence, with epidemiological data suggesting that some populations have a higher risk of developing this disease. However, to date, most genetic studies of type 2 diabetes and related glycaemic traits have been performed in individuals of European ancestry. The same is true for most other complex diseases, largely due to use of 'convenience samples'. Rapid genotyping of large population cohorts and case-control studies from existing collections was performed when the genome-wide association study (GWAS) 'revolution' began, back in 2005. Although global representation has increased in the intervening 15 years, further expansion and inclusion of diverse populations in genetic and genomic studies is still needed. In this review, I discuss the progress made in incorporating multi-ancestry participants in genetic analyses of type 2 diabetes and related glycaemic traits, and associated opportunities and challenges. I also discuss how increased representation of global diversity in genetic and genomic studies is required to fulfil the promise of precision medicine for all.
Collapse
Affiliation(s)
- Inês Barroso
- Exeter Centre of Excellence for Diabetes research (EXCEED), University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
21
|
Qu J, Qu HQ, Bradfield JP, Glessner JT, Chang X, Tian L, March M, Connolly JJ, Roizen JD, Sleiman PMA, Hakonarson H. Insights into non-autoimmune type 1 diabetes with 13 novel loci in low polygenic risk score patients. Sci Rep 2021; 11:16013. [PMID: 34362956 PMCID: PMC8346538 DOI: 10.1038/s41598-021-94994-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/20/2021] [Indexed: 01/21/2023] Open
Abstract
With polygenic risk score (PRS) for autoimmune type 1 diabetes (T1D), this study identified T1D cases with low T1D PRS and searched for susceptibility loci in these cases. Our hypothesis is that genetic effects (likely mediated by relatively rare genetic variants) of non-mainstream (or non-autoimmune) T1D might have been diluted in the previous studies on T1D cases in general. Two cohorts for the PRS modeling and testing respectively were included. The first cohort consisted of 3302 T1D cases and 6181 controls, and the independent second cohort consisted of 3297 T1D cases and 6169 controls. Cases with low T1D PRS were identified using PRSice-2 and compared to controls with low T1D PRS by genome-wide association (GWA) test. Thirteen novel genetic loci with high imputation quality (Quality Score r2 > 0.91) were identified of SNPs/SNVs associated with low PRS T1D at genome-wide significance (P ≤ 5.0 × E-08), in addition to 4 established T1D loci, 3 reported loci by our previous study, as well as 9 potential novel loci represented by rare SNVs, but with relatively low imputation quality (Quality Score r2 < 0.90). For the 13 novel loci, 9 regions have been reported of association with obesity related traits by previous GWA studies. Three loci encoding long intergenic non-protein coding RNAs (lncRNA), and 2 loci involved in N-linked glycosylation are also highlighted in this study.
Collapse
Affiliation(s)
- Jingchun Qu
- grid.239552.a0000 0001 0680 8770The Center for Applied Genomics, Children’s Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA 19104 USA
| | - Hui-Qi Qu
- grid.239552.a0000 0001 0680 8770The Center for Applied Genomics, Children’s Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA 19104 USA
| | | | - Joseph T. Glessner
- grid.239552.a0000 0001 0680 8770The Center for Applied Genomics, Children’s Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA 19104 USA
| | - Xiao Chang
- grid.239552.a0000 0001 0680 8770The Center for Applied Genomics, Children’s Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA 19104 USA
| | - Lifeng Tian
- grid.239552.a0000 0001 0680 8770The Center for Applied Genomics, Children’s Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA 19104 USA
| | - Michael March
- grid.239552.a0000 0001 0680 8770The Center for Applied Genomics, Children’s Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA 19104 USA
| | - John J. Connolly
- grid.239552.a0000 0001 0680 8770The Center for Applied Genomics, Children’s Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA 19104 USA
| | - Jeffrey D. Roizen
- grid.25879.310000 0004 1936 8972Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Patrick M. A. Sleiman
- grid.239552.a0000 0001 0680 8770The Center for Applied Genomics, Children’s Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA 19104 USA ,grid.25879.310000 0004 1936 8972Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
| | - Hakon Hakonarson
- grid.239552.a0000 0001 0680 8770The Center for Applied Genomics, Children’s Hospital of Philadelphia, 3615 Civic Center Blvd, Abramson Building, Philadelphia, PA 19104 USA ,grid.25879.310000 0004 1936 8972Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA ,grid.239552.a0000 0001 0680 8770Division of Pulmonary Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
| |
Collapse
|
22
|
Reid S. From Prehistory to Precision Medicine: Are Genetic Risk Scores Our Next Step? J Rheumatol 2021; 48:799-801. [PMID: 33722950 DOI: 10.3899/jrheum.201256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Sarah Reid
- S. Reid, PhD Student, MD, Rheumatology and Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
23
|
Zhang H, Colclough K, Gloyn AL, Pollin TI. Monogenic diabetes: a gateway to precision medicine in diabetes. J Clin Invest 2021; 131:142244. [PMID: 33529164 PMCID: PMC7843214 DOI: 10.1172/jci142244] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Monogenic diabetes refers to diabetes mellitus (DM) caused by a mutation in a single gene and accounts for approximately 1%-5% of diabetes. Correct diagnosis is clinically critical for certain types of monogenic diabetes, since the appropriate treatment is determined by the etiology of the disease (e.g., oral sulfonylurea treatment of HNF1A/HNF4A-diabetes vs. insulin injections in type 1 diabetes). However, achieving a correct diagnosis requires genetic testing, and the overlapping of the clinical features of monogenic diabetes with those of type 1 and type 2 diabetes has frequently led to misdiagnosis. Improvements in sequencing technology are increasing opportunities to diagnose monogenic diabetes, but challenges remain. In this Review, we describe the types of monogenic diabetes, including common and uncommon types of maturity-onset diabetes of the young, multiple causes of neonatal DM, and syndromic diabetes such as Wolfram syndrome and lipodystrophy. We also review methods of prioritizing patients undergoing genetic testing, and highlight existing challenges facing sequence data interpretation that can be addressed by forming collaborations of expertise and by pooling cases.
Collapse
Affiliation(s)
- Haichen Zhang
- University of Maryland School of Medicine, Department of Medicine, Baltimore, Maryland, USA
| | - Kevin Colclough
- Exeter Genomics Laboratory, Royal Devon and Exeter Hospital, Exeter, United Kingdom
| | - Anna L. Gloyn
- Department of Pediatrics, Division of Endocrinology, and,Stanford Diabetes Research Center, Stanford School of Medicine, Stanford, California, USA
| | - Toni I. Pollin
- University of Maryland School of Medicine, Department of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
24
|
Johnson MB, Hattersley AT, Patel KA. More on STAT1 Gain of Function, Type 1 Diabetes, and JAK Inhibition. N Engl J Med 2021; 384:93-94. [PMID: 33406351 DOI: 10.1056/nejmc2032806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
25
|
Amanat S, Requena T, Lopez-Escamez JA. A Systematic Review of Extreme Phenotype Strategies to Search for Rare Variants in Genetic Studies of Complex Disorders. Genes (Basel) 2020; 11:genes11090987. [PMID: 32854191 PMCID: PMC7564972 DOI: 10.3390/genes11090987] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
Exome sequencing has been commonly used to characterize rare diseases by selecting multiplex families or singletons with an extreme phenotype (EP) and searching for rare variants in coding regions. The EP strategy covers both extreme ends of a disease spectrum and it has been also used to investigate the contribution of rare variants to the heritability of complex clinical traits. We conducted a systematic review to find evidence supporting the use of EP strategies in the search for rare variants in genetic studies of complex diseases and highlight the contribution of rare variations to the genetic structure of polygenic conditions. After assessing the quality of the retrieved records, we selected 19 genetic studies considering EPs to demonstrate genetic association. All studies successfully identified several rare or de novo variants, and many novel candidate genes were also identified by selecting an EP. There is enough evidence to support that the EP approach for patients with an early onset of a disease can contribute to the identification of rare variants in candidate genes or pathways involved in complex diseases. EP patients may contribute to a better understanding of the underlying genetic architecture of common heterogeneous disorders such as tinnitus or age-related hearing loss.
Collapse
Affiliation(s)
- Sana Amanat
- Otology & Neurotology Group CTS495, Department of Genomic Medicine, GENYO—Centre for Genomics and Oncological Research—Pfizer/University of Granada/Junta de Andalucía, PTS, 18016 Granada, Spain;
| | - Teresa Requena
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9JZ, UK;
| | - Jose Antonio Lopez-Escamez
- Otology & Neurotology Group CTS495, Department of Genomic Medicine, GENYO—Centre for Genomics and Oncological Research—Pfizer/University of Granada/Junta de Andalucía, PTS, 18016 Granada, Spain;
- Department of Otolaryngology, Instituto de Investigación Biosanitaria ibs.GRANADA, Hospital Universitario Virgen de las Nieves, Universidad de Granada, 18016 Granada, Spain
- Department of Surgery, Division of Otolaryngology, Universidad de Granada, 18016 Granada, Spain
- Correspondence: ; Tel.: +34-958-715-500-160
| |
Collapse
|
26
|
Yaghootkar H, Abbasi F, Ghaemi N, Rabbani A, Wakeling MN, Eshraghi P, Enayati S, Vakili S, Heidari S, Patel K, Sayarifard F, Borhan‐Dayani S, McDonald TJ, Ellard S, Hattersley AT, Amoli MM, Vakili R, Colclough K. Type 1 diabetes genetic risk score discriminates between monogenic and Type 1 diabetes in children diagnosed at the age of <5 years in the Iranian population. Diabet Med 2019; 36:1694-1702. [PMID: 31276222 PMCID: PMC7027759 DOI: 10.1111/dme.14071] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/03/2019] [Indexed: 12/24/2022]
Abstract
AIM To examine the extent to which discriminatory testing using antibodies and Type 1 diabetes genetic risk score, validated in European populations, is applicable in a non-European population. METHODS We recruited 127 unrelated children with diabetes diagnosed between 9 months and 5 years from two centres in Iran. All children underwent targeted next-generation sequencing of 35 monogenic diabetes genes. We measured three islet autoantibodies (islet antigen 2, glutamic acid decarboxylase and zinc transporter 8) and generated a Type 1 diabetes genetic risk score in all children. RESULTS We identified six children with monogenic diabetes, including four novel mutations: homozygous mutations in WFS1 (n=3), SLC19A2 and SLC29A3, and a heterozygous mutation in GCK. All clinical features were similar in children with monogenic diabetes (n=6) and in the rest of the cohort (n=121). The Type 1 diabetes genetic risk score discriminated children with monogenic from Type 1 diabetes [area under the receiver-operating characteristic curve 0.90 (95% CI 0.83-0.97)]. All children with monogenic diabetes were autoantibody-negative. In children with no mutation, 59 were positive to glutamic acid decarboxylase, 39 to islet antigen 2 and 31 to zinc transporter 8. Measuring zinc transporter 8 increased the number of autoantibody-positive individuals by eight. CONCLUSIONS The present study provides the first evidence that Type 1 diabetes genetic risk score can be used to distinguish monogenic from Type 1 diabetes in an Iranian population with a large number of consanguineous unions. This test can be used to identify children with a higher probability of having monogenic diabetes who could then undergo genetic testing. Identification of these individuals would reduce the cost of treatment and improve the management of their clinical course.
Collapse
Affiliation(s)
- H. Yaghootkar
- Genetics of Complex TraitsUniversity of Exeter Medical School, Royal Devon & Exeter HospitalExeterUK
| | - F. Abbasi
- Growth and Development Research CentreTehran University of Medical SciencesTehranIran
| | - N. Ghaemi
- Department of Paediatric DiseaseFaulty of Medicine, Mashhad University of Medical SciencesMashhadIran
| | - A. Rabbani
- Growth and Development Research CentreTehran University of Medical SciencesTehranIran
| | - M. N. Wakeling
- Institute of Biomedical and Clinical ScienceUniversity of Exeter Medical SchoolExeterUK
| | - P. Eshraghi
- Department of Paediatric DiseaseFaulty of Medicine, Mashhad University of Medical SciencesMashhadIran
| | - S. Enayati
- Metabolic Disorders Research CentreEndocrinology and Metabolism Molecular-Cellular Sciences InstituteTehran University of Medical SciencesTehranIran
| | - S. Vakili
- Medical Genetics Research CentreMashhad University of Medical SciencesMashhadIran
| | - S. Heidari
- Growth and Development Research CentreTehran University of Medical SciencesTehranIran
| | - K. Patel
- Institute of Biomedical and Clinical ScienceUniversity of Exeter Medical SchoolExeterUK
| | - F. Sayarifard
- Growth and Development Research CentreTehran University of Medical SciencesTehranIran
| | - S. Borhan‐Dayani
- Metabolic Disorders Research CentreEndocrinology and Metabolism Molecular-Cellular Sciences InstituteTehran University of Medical SciencesTehranIran
| | - T. J. McDonald
- Institute of Biomedical and Clinical ScienceUniversity of Exeter Medical SchoolExeterUK
- Departments of Clinical BiochemistryRoyal Devon and Exeter NHS Foundation TrustExeterUK
| | - S. Ellard
- Institute of Biomedical and Clinical ScienceUniversity of Exeter Medical SchoolExeterUK
- Departments of Clinical BiochemistryRoyal Devon and Exeter NHS Foundation TrustExeterUK
| | - A. T. Hattersley
- Institute of Biomedical and Clinical ScienceUniversity of Exeter Medical SchoolExeterUK
| | - M. M. Amoli
- Metabolic Disorders Research CentreEndocrinology and Metabolism Molecular-Cellular Sciences InstituteTehran University of Medical SciencesTehranIran
| | - R. Vakili
- Department of Paediatric DiseaseFaulty of Medicine, Mashhad University of Medical SciencesMashhadIran
- Medical Genetics Research CentreMashhad University of Medical SciencesMashhadIran
| | - K. Colclough
- Departments of Molecular GeneticsRoyal Devon and Exeter NHS Foundation TrustExeterUK
| |
Collapse
|
27
|
Udler MS, McCarthy MI, Florez JC, Mahajan A. Genetic Risk Scores for Diabetes Diagnosis and Precision Medicine. Endocr Rev 2019; 40:1500-1520. [PMID: 31322649 PMCID: PMC6760294 DOI: 10.1210/er.2019-00088] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022]
Abstract
During the last decade, there have been substantial advances in the identification and characterization of DNA sequence variants associated with individual predisposition to type 1 and type 2 diabetes. As well as providing insights into the molecular, cellular, and physiological mechanisms involved in disease pathogenesis, these risk variants, when combined into a polygenic score, capture information on individual patterns of disease predisposition that have the potential to influence clinical management. In this review, we describe the various opportunities that polygenic scores provide: to predict diabetes risk, to support differential diagnosis, and to understand phenotypic and clinical heterogeneity. We also describe the challenges that will need to be overcome if this potential is to be fully realized.
Collapse
Affiliation(s)
- Miriam S Udler
- Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Headington, Oxford, United Kingdom
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jose C Florez
- Diabetes Unit, Massachusetts General Hospital, Boston, Massachusetts
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Anubha Mahajan
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
28
|
Strakova V, Elblova L, Johnson MB, Dusatkova P, Obermannova B, Petruzelkova L, Kolouskova S, Snajderova M, Fronkova E, Svaton M, Lebl J, Hattersley AT, Sumnik Z, Pruhova S. Screening of monogenic autoimmune diabetes among children with type 1 diabetes and multiple autoimmune diseases: is it worth doing? J Pediatr Endocrinol Metab 2019; 32:1147-1153. [PMID: 31483759 DOI: 10.1515/jpem-2019-0261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 08/09/2019] [Indexed: 11/15/2022]
Abstract
Background Paediatric type 1 diabetes (T1D) and rare syndromes of monogenic multi-organ autoimmunity share basic features such as full insulin dependency and the presence of circulating beta-cell autoantibodies. However, the aetiopathogenesis, natural course and treatment of these conditions differ; therefore, monogenic multi-organ autoimmunity requires early recognition. We aimed to search for these monogenic conditions among a large cohort of children with T1D. Methods Of 519 children with T1D followed-up in a single centre, 18 had multiple additional autoimmune conditions - either autoimmune thyroid disease (AITD) and coeliac disease (CD) or at least one additional organ-specific autoimmune condition in addition to AITD or CD. These 18 children were tested by direct Sanger sequencing (four patients with a suggestive phenotype of immune dysregulation, polyendocrinopathy, enteropathy, X-linked [IPEX] or signal transducer and activator of transcription 3 [STAT3]- and cytotoxic T-lymphocyte protein 4 [CTLA4]-associated syndromes) or by whole-exome sequencing (WES) focused on autoimmune regulator (AIRE), forkhead box protein 3 (FOXP3), CTLA4, STAT3, signal transducer and activator of transcription 1 (STAT1), lipopolysaccharide-responsive and beige-like anchor protein (LRBA) and interleukin-2 receptor subunit α (IL2RA) genes. In addition, we assessed their T1D genetic risk score (T1D-GRS). Results We identified novel variants in FOXP3, STAT3 and CTLA4 in four cases. All patients had a severe phenotype suggestive of a single gene defect. No variants were identified in the remaining 14 patients. T1D-GRS varied among the entire cohort; four patients had scores below the 25th centile including two genetically confirmed cases. Conclusions A monogenic cause of autoimmune diabetes was confirmed only in four patients. Genetic screening for monogenic autoimmunity in children with a milder phenotype and a combination of AITD and CD is unlikely to identify a monogenic cause. In addition, the T1D-GRS varied among individual T1D patients.
Collapse
Affiliation(s)
- Veronika Strakova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Lenka Elblova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Matthew B Johnson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Petra Dusatkova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Barbora Obermannova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Lenka Petruzelkova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Stanislava Kolouskova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Marta Snajderova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Eva Fronkova
- Department of Paediatric Haematology and Oncology, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Michael Svaton
- Department of Paediatric Haematology and Oncology, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Jan Lebl
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Andrew T Hattersley
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Zdenek Sumnik
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| | - Stepanka Pruhova
- Department of Paediatrics, 2Faculty of Medicine, Charles University in Prague, University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
29
|
De Franco E. From Biology to Genes and Back Again: Gene Discovery for Monogenic Forms of Beta-Cell Dysfunction in Diabetes. J Mol Biol 2019; 432:1535-1550. [PMID: 31479665 DOI: 10.1016/j.jmb.2019.08.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/22/2019] [Accepted: 08/22/2019] [Indexed: 12/14/2022]
Abstract
This review focuses on gene discovery strategies used to identify monogenic forms of diabetes caused by reduced pancreatic beta-cell number (due to destruction or defective development) or impaired beta-cell function. Gene discovery efforts in monogenic diabetes have identified 36 genes so far. These genetic causes have been identified using four main approaches: linkage analysis, candidate gene sequencing and most recently, exome and genome sequencing. The advent of next-generation sequencing has allowed researchers to move away from linkage analysis (relying on large pedigrees and/or multiple families with the same genetic condition) and candidate gene (relying on previous knowledge on the gene's role) strategies to use a gene agnostic approach, utilizing genetic evidence (such as variant frequency, predicted variant effect on protein function, and predicted mode of inheritance) to identify the causative mutation. This approach led to the identification of seven novel genetic causes of monogenic diabetes, six by exome sequencing and one by genome sequencing. In many of these cases, the disease-causing gene was not known to be important for beta-cell function prior to the gene discovery study. These novel findings highlight a new role for gene discovery studies in furthering our understanding of beta-cell function and dysfunction in diabetes. While many gene discovery studies in the past were led by knowledge in the field (through the candidate gene strategy), now they often lead the scientific advances in the field by identifying new important biological players to be further characterized by in vitro and in vivo studies.
Collapse
Affiliation(s)
- Elisa De Franco
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, EX2 5DW Exeter, UK; Institute of Biomedical and Clinical Science, Level 3, RILD Building, Barrack Road, EX2 5DW Exeter, United Kingdom.
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW The genetic risk for type 1 diabetes has been studied for over half a century, with the strong genetic associations of type 1 diabetes forming critical evidence for the role of the immune system in pathogenesis. In this review, we discuss some of the original research leading to recent developments in type 1 diabetes genetics. RECENT FINDINGS We examine the translation of polygenic scores for type 1 diabetes into tools for prediction and diagnosis of type 1 diabetes, in particular, when used in combination with other biomarkers and clinical features, such as age and islet-specific autoantibodies. Furthermore, we review the description of age associations with type 1 diabetes genetic risk, and the investigation of loci linked to type 2 diabetes in progression of type 1 diabetes. Finally, we consider current limitations, including the scarcity of data from racial and ethnic minorities, and future directions. SUMMARY The development of polygenic risk scores has allowed the integration of type 1 diabetes genetics into diagnosis and prediction. Emerging information on the role of specific genes in subgroups of individuals with the disease, for example, early-onset, mild autoimmunity, and so forth, is facilitating our understanding of the heterogeneity of type 1 diabetes, with the ultimate goal of using genetic information in research and clinical practice.
Collapse
Affiliation(s)
- Richard A Oram
- RILD Level 3, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Royal Devon and Exeter Hospital
- NIHR Exeter Clinical Research Facility, University of Exeter Medical School
- The Academic Renal Unit, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Maria J Redondo
- Pediatric Diabetes and Endocrinology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
31
|
Shapiro MR, Atkinson MA, Brusko TM. Pleiotropic roles of the insulin-like growth factor axis in type 1 diabetes. Curr Opin Endocrinol Diabetes Obes 2019; 26:188-194. [PMID: 31145130 PMCID: PMC7135378 DOI: 10.1097/med.0000000000000484] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW We review studies demonstrating lowered levels of insulin-like growth factors (IGFs) in patients with recent-onset type 1 diabetes (T1D) and discuss their potential roles in the disorder's pathogenesis. RECENT FINDINGS IGFs have long been recognized as a class of hormones that promote growth, development, and cellular metabolism throughout the human body. More recently, studies have noted an association between reduced pancreatic weight/volume and T1D. Thus, we believe it is important to understand pancreatic regulation of IGF expression and bioavailability, as well as the impact of IGFs on pancreatic growth and islet health. Additional studies of IGFs have been extended to their influence on the inflammatory/regulatory balance of monocytes, B cells, and T cells; features which have been previously established to show dysregulation in settings of T1D. SUMMARY These data suggest that IGFs may prevent known impairments in the pancreas and immune system in T1D and underscore the need to extend these studies, some of which were performed in health or other autoimmune diseases, toward T1D specifically. Collectively, the work emphasized here support the potential therapeutic use of IGFs in T1D prevention efforts as pancreatic growth factors and/or immunoregulatory agents.
Collapse
Affiliation(s)
- Melanie R. Shapiro
- Department of Pathology, Immunology, and Laboratory Medicine, The University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Mark A. Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, The University of Florida Diabetes Institute, Gainesville, Florida, USA
- Department of Pediatrics, The University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Todd M. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, The University of Florida Diabetes Institute, Gainesville, Florida, USA
| |
Collapse
|
32
|
What is new in HIES? Recent insights from the interface of primary immune deficiency and atopy. Curr Opin Allergy Clin Immunol 2019; 18:445-452. [PMID: 30188342 DOI: 10.1097/aci.0000000000000481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Understanding the pathophysiology of monogenic primary immunodeficiency (PID) with atopic presentation has pivotal implications for intervention strategies and potentially wider polygenic atopic-related traits. This review will discuss advances in gene discovery arising from monogenic defects at the interface between PID and atopy, notably the hyper-IgE syndromes. RECENT FINDINGS Key molecular pathways underlying development of primary atopic diseases have recently been proposed. We test this classification through reviewing novel genes reported in the last 2 years and compare insights from pathway-analysis of genome-wide association studies (GWAS) of atopic-related traits.Growing access to next-generation sequencing (NGS) has resulted in a surge in gene discovery, highlighting the utility and some pitfalls of this approach in clinical practice. The variability of presenting phenotypes reveals important gene-dosage effects. This has important implications for therapeutic strategies such as protein stabilization and modulators of JAK-STAT or TH2-cytokine signalling. We also consider the therapeutic implications raised by CARD11 deficiency, and wider applications of NGS including polygenic risk score in atopy. SUMMARY Disorders presenting at the interface between PID and allergy are often difficult to diagnose, with serious consequences if missed. Application of NGS has already provided critical insights to pathways enabling targeted therapeutic interventions, and potential wider translation to polygenic disorders.
Collapse
|
33
|
Johnson MB, De Franco E, Greeley SAW, Letourneau LR, Gillespie KM, Wakeling MN, Ellard S, Flanagan SE, Patel KA, Hattersley AT. Trisomy 21 Is a Cause of Permanent Neonatal Diabetes That Is Autoimmune but Not HLA Associated. Diabetes 2019; 68:1528-1535. [PMID: 30962220 PMCID: PMC6609990 DOI: 10.2337/db19-0045] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/28/2019] [Indexed: 02/07/2023]
Abstract
Identifying new causes of permanent neonatal diabetes (PNDM) (diagnosis <6 months) provides important insights into β-cell biology. Patients with Down syndrome (DS) resulting from trisomy 21 are four times more likely to have childhood diabetes with an intermediate HLA association. It is not known whether DS can cause PNDM. We found that trisomy 21 was seven times more likely in our PNDM cohort than in the population (13 of 1,522 = 85 of 10,000 observed vs. 12.6 of 10,000 expected) and none of the 13 DS-PNDM patients had a mutation in the known PNDM genes that explained 82.9% of non-DS PNDM. Islet autoantibodies were present in 4 of 9 DS-PNDM patients, but DS-PNDM was not associated with polygenic susceptibility to type 1 diabetes (T1D). We conclude that trisomy 21 is a cause of autoimmune PNDM that is not HLA associated. We propose that autoimmune diabetes in DS is heterogeneous and includes coincidental T1D that is HLA associated and diabetes caused by trisomy 21 that is not HLA associated.
Collapse
Affiliation(s)
- Matthew B Johnson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Elisa De Franco
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Siri Atma W Greeley
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, The University of Chicago, Chicago, IL
| | - Lisa R Letourneau
- Kovler Diabetes Center, Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, The University of Chicago, Chicago, IL
| | | | - Matthew N Wakeling
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Sian Ellard
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Sarah E Flanagan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Kashyap A Patel
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Andrew T Hattersley
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, U.K.
| |
Collapse
|
34
|
Fabbri M, Frixou M, Degano M, Fousteri G. Type 1 Diabetes in STAT Protein Family Mutations: Regulating the Th17/Treg Equilibrium and Beyond. Diabetes 2019; 68:258-265. [PMID: 30665954 DOI: 10.2337/db18-0627] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 11/11/2018] [Indexed: 11/13/2022]
Abstract
Improvements in the immunological, molecular, and genetic technologies such as next-generation sequencing have led to an exponential increase in the number of monogenic immune dysregulatory syndromes diagnosed, where type 1 diabetes (T1D) forms part of the autoimmune manifestations. Here, we reviewed the mutations in the signal transducer and activator of transcription (STAT) protein family, namely gain-of-function (GOF) mutations in STAT1 and STAT3 as well as STAT5b deficiency, that show strong association to T1D susceptibility. The equilibrium of T-helper 17 (Th17) and regulatory T cells (Tregs) is often found altered in patients affected by STAT GOF mutations. While the increased number of Th17 cells and the concomitant decrease in Treg cells may explain T1D in STAT3 GOF patients, the reduced number of Th17 cells found in those carrying STAT1 GOF mutations added a new level of complexity on the exact role of Th17 in the pathogenesis of T1D. Here, we describe the possible mechanisms through which STAT3 and STAT1 GOF mutations may perturb the fate and function of Th17 and Tregs and explore how this may lead to the development of T1D. We propose that the study of monogenic diseases, and in particular STAT mutations, may not only improve our understanding of the function of the human immune system but also shed light onto the pathogenic mechanisms of T1D and the genetic variants that confer predisposition to the disease.
Collapse
Affiliation(s)
- Marco Fabbri
- Division of Immunology, Transplantation and Infectious Diseases, Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Mikaela Frixou
- School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, U.K
| | - Massimo Degano
- Biocrystallography Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Georgia Fousteri
- Division of Immunology, Transplantation and Infectious Diseases, Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Monogenic forms of diabetes have received increased attention and genetic testing is more widely available; however, many patients are still misdiagnosed as having type 1 (T1D) or type 2 diabetes. This review will address updates to monogenic diabetes prevalence, identification, treatment, and genetic testing. RECENT FINDINGS The creation of a T1D genetic risk score and the use of noninvasive urinary C-peptide creatinine ratios have provided new tools to aid in the discrimination of possible monogenic diabetes from likely T1D. Early, high-dose sulfonylurea treatment in infants with a KCNJ11 or ABCC8 mutation continues to be well tolerated and effective. As the field moves towards more comprehensive genetic testing methods, there is an increased opportunity to identify novel genetic causes. Genetic testing results continue to allow for personalized treatment but should provide patient information at an appropriate health literacy level. SUMMARY Although there have been clinical and genetic advances in monogenic diabetes, patients are still misdiagnosed. Improved insurance coverage of genetic testing is needed. The majority of data on monogenic diabetes has been collected from Caucasian populations, therefore, research studies should endeavor to include broader ethnic and racial diversity to provide comprehensive information for all populations.
Collapse
|
36
|
Unravelling the Roles of Susceptibility Loci for Autoimmune Diseases in the Post-GWAS Era. Genes (Basel) 2018; 9:genes9080377. [PMID: 30060490 PMCID: PMC6115971 DOI: 10.3390/genes9080377] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/06/2018] [Accepted: 07/23/2018] [Indexed: 12/18/2022] Open
Abstract
Although genome-wide association studies (GWAS) have identified several hundred loci associated with autoimmune diseases, their mechanistic insights are still poorly understood. The human genome is more complex than single nucleotide polymorphisms (SNPs) that are interrogated by GWAS arrays. Apart from SNPs, it also comprises genetic variations such as insertions-deletions, copy number variations, and somatic mosaicism. Although previous studies suggest that common copy number variations do not play a major role in autoimmune disease risk, it is possible that certain rare genetic variations with large effect sizes are relevant to autoimmunity. In addition, other layers of regulations such as gene-gene interactions, epigenetic-determinants, gene and environmental interactions also contribute to the heritability of autoimmune diseases. This review focuses on discussing why studying these elements may allow us to gain a more comprehensive understanding of the aetiology of complex autoimmune traits.
Collapse
|
37
|
Letourneau LR, Greeley SAW. Congenital Diabetes: Comprehensive Genetic Testing Allows for Improved Diagnosis and Treatment of Diabetes and Other Associated Features. Curr Diab Rep 2018; 18:46. [PMID: 29896650 PMCID: PMC6341981 DOI: 10.1007/s11892-018-1016-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW The goal of this review is to provide updates on congenital (neonatal) diabetes from 2011 to present, with an emphasis on publications from 2015 to present. RECENT FINDINGS There has been continued worldwide progress in uncovering the genetic causes of diabetes presenting within the first year of life, including the recognition of nine new causes since 2011. Management has continued to be refined based on underlying molecular cause, and longer-term experience has provided better understanding of the effectiveness, safety, and sustainability of treatment. Associated conditions have been further clarified, such as neurodevelopmental delays and pancreatic insufficiency, including a better appreciation for how these "secondary" conditions impact quality of life for patients and their families. While continued research is essential to understand all forms of congenital diabetes, these cases remain a compelling example of personalized genetic medicine.
Collapse
Affiliation(s)
- Lisa R Letourneau
- Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Kovler Diabetes Center, The University of Chicago, MC 1027/N235; 5841 S. Maryland Ave., Chicago, IL, 60637, USA
| | - Siri Atma W Greeley
- Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Kovler Diabetes Center, The University of Chicago, MC 1027/N235; 5841 S. Maryland Ave., Chicago, IL, 60637, USA.
| |
Collapse
|