1
|
Sarkar S, Zheng X, Clair GC, Kwon YM, You Y, Swensen AC, Webb-Robertson BJM, Nakayasu ES, Qian WJ, Metz TO. Exploring new frontiers in type 1 diabetes through advanced mass-spectrometry-based molecular measurements. Trends Mol Med 2024; 30:1137-1151. [PMID: 39152082 PMCID: PMC11631641 DOI: 10.1016/j.molmed.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/19/2024]
Abstract
Type 1 diabetes (T1D) is a devastating autoimmune disease for which advanced mass spectrometry (MS) methods are increasingly used to identify new biomarkers and better understand underlying mechanisms. For example, integration of MS analysis and machine learning has identified multimolecular biomarker panels. In mechanistic studies, MS has contributed to the discovery of neoepitopes, and pathways involved in disease development and identifying therapeutic targets. However, challenges remain in understanding the role of tissue microenvironments, spatial heterogeneity, and environmental factors in disease pathogenesis. Recent advancements in MS, such as ultra-fast ion-mobility separations, and single-cell and spatial omics, can play a central role in addressing these challenges. Here, we review recent advancements in MS-based molecular measurements and their role in understanding T1D.
Collapse
Affiliation(s)
- Soumyadeep Sarkar
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Xueyun Zheng
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Geremy C Clair
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Yu Mi Kwon
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Youngki You
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | - Adam C Swensen
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA
| | | | - Ernesto S Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA.
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA.
| | - Thomas O Metz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99352, USA.
| |
Collapse
|
2
|
Stephens SB. Defining Cytokine Responsive and Non-responsive Human β-cells. FUNCTION 2024; 5:zqae039. [PMID: 39251388 PMCID: PMC11406544 DOI: 10.1093/function/zqae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024] Open
Affiliation(s)
- Samuel B Stephens
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52242, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA, 52242, USA
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
3
|
Cabiati M, Federico G, Del Ry S. Importance of Studying Non-Coding RNA in Children and Adolescents with Type 1 Diabetes. Biomedicines 2024; 12:1988. [PMID: 39335501 PMCID: PMC11429055 DOI: 10.3390/biomedicines12091988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Type 1 diabetes (T1D) mellitus is a chronic illness in children and teens, with rising global incidence rates. It stems from an autoimmune attack on pancreatic β cells, leading to insufficient insulin production. Genetic susceptibility and environmental triggers initiate this process. Early detection is possible by identifying multiple autoantibodies, which aids in predicting future T1D development. A new staging system highlights T1D's onset with islet autoimmunity rather than symptoms. Family members of T1D patients face a significantly increased risk of T1D. Italy recently passed a law mandating national T1D screening for pediatric populations. Measurements of β cell function continue to be essential in assessing efficacy, and different models have been proposed, but more appropriate biomarkers are mandatory for both progression studies before the onset of diabetes and during therapeutic monitoring. Biomarkers like microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) play key roles in T1D pathogenesis by regulating gene expression. Understanding their roles offers insights into T1D mechanisms and potential therapeutic targets. In this review, we summarized recent progress in the roles of some non-coding RNAs (ncRNAs) in the pathogenesis of T1D, with particular attention to miRNAs, lncRNAs, and circRNAs.
Collapse
Affiliation(s)
- Manuela Cabiati
- Laboratory of Biochemistry and Molecular Biology, Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | - Giovanni Federico
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Silvia Del Ry
- Laboratory of Biochemistry and Molecular Biology, Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| |
Collapse
|
4
|
Phillip M, Achenbach P, Addala A, Albanese-O'Neill A, Battelino T, Bell KJ, Besser REJ, Bonifacio E, Colhoun HM, Couper JJ, Craig ME, Danne T, de Beaufort C, Dovc K, Driscoll KA, Dutta S, Ebekozien O, Larsson HE, Feiten DJ, Frohnert BI, Gabbay RA, Gallagher MP, Greenbaum CJ, Griffin KJ, Hagopian W, Haller MJ, Hendrieckx C, Hendriks E, Holt RIG, Hughes L, Ismail HM, Jacobsen LM, Johnson SB, Kolb LE, Kordonouri O, Lange K, Lash RW, Lernmark Å, Libman I, Lundgren M, Maahs DM, Marcovecchio ML, Mathieu C, Miller KM, O'Donnell HK, Oron T, Patil SP, Pop-Busui R, Rewers MJ, Rich SS, Schatz DA, Schulman-Rosenbaum R, Simmons KM, Sims EK, Skyler JS, Smith LB, Speake C, Steck AK, Thomas NPB, Tonyushkina KN, Veijola R, Wentworth JM, Wherrett DK, Wood JR, Ziegler AG, DiMeglio LA. Consensus guidance for monitoring individuals with islet autoantibody-positive pre-stage 3 type 1 diabetes. Diabetologia 2024; 67:1731-1759. [PMID: 38910151 PMCID: PMC11410955 DOI: 10.1007/s00125-024-06205-5] [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] [Indexed: 06/25/2024]
Abstract
Given the proven benefits of screening to reduce diabetic ketoacidosis (DKA) likelihood at the time of stage 3 type 1 diabetes diagnosis, and emerging availability of therapy to delay disease progression, type 1 diabetes screening programmes are being increasingly emphasised. Once broadly implemented, screening initiatives will identify significant numbers of islet autoantibody-positive (IAb+) children and adults who are at risk of (confirmed single IAb+) or living with (multiple IAb+) early-stage (stage 1 and stage 2) type 1 diabetes. These individuals will need monitoring for disease progression; much of this care will happen in non-specialised settings. To inform this monitoring, JDRF in conjunction with international experts and societies developed consensus guidance. Broad advice from this guidance includes the following: (1) partnerships should be fostered between endocrinologists and primary-care providers to care for people who are IAb+; (2) when people who are IAb+ are initially identified there is a need for confirmation using a second sample; (3) single IAb+ individuals are at lower risk of progression than multiple IAb+ individuals; (4) individuals with early-stage type 1 diabetes should have periodic medical monitoring, including regular assessments of glucose levels, regular education about symptoms of diabetes and DKA, and psychosocial support; (5) interested people with stage 2 type 1 diabetes should be offered trial participation or approved therapies; and (6) all health professionals involved in monitoring and care of individuals with type 1 diabetes have a responsibility to provide education. The guidance also emphasises significant unmet needs for further research on early-stage type 1 diabetes to increase the rigour of future recommendations and inform clinical care.
Collapse
Affiliation(s)
- Moshe Phillip
- Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Ananta Addala
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Tadej Battelino
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Endocrinology, Diabetes and Metabolism, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Kirstine J Bell
- Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Rachel E J Besser
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre Human Genetics, Nuffield Department of Medicine Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technical University of Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, Helmholtz Centre Munich at the University Clinic Carl Gustav Carus of TU Dresden and Faculty of Medicine, Dresden, Germany
| | - Helen M Colhoun
- The Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Department of Public Health, NHS Fife, Kirkcaldy, UK
| | - Jennifer J Couper
- Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Division of Paediatrics, Women's and Children's Hospital, Adelaide, SA, Australia
| | - Maria E Craig
- Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Discipline of Paediatrics & Child Health, School of Clinical Medicine, UNSW Medicine & Health, Sydney, NSW, Australia
| | | | - Carine de Beaufort
- International Society for Pediatric and Adolescent Diabetes (ISPAD), Berlin, Germany
- Diabetes & Endocrine Care Clinique Pédiatrique (DECCP), Clinique Pédiatrique/Centre Hospitalier (CH) de Luxembourg, Luxembourg City, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-Belval, Luxembourg
| | - Klemen Dovc
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Endocrinology, Diabetes and Metabolism, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Kimberly A Driscoll
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
- Department of Pediatrics, University of Florida Diabetes Institute, Gainesville, FL, USA
| | | | | | - Helena Elding Larsson
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Pediatrics, Skåne University Hospital, Malmö and Lund, Sweden
| | | | - Brigitte I Frohnert
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Carla J Greenbaum
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, WA, USA
| | - Kurt J Griffin
- Sanford Research, Sioux Falls, SD, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
| | - William Hagopian
- Pacific Northwest Diabetes Research Institute, University of Washington, Seattle, WA, USA
| | - Michael J Haller
- Department of Pediatrics, University of Florida Diabetes Institute, Gainesville, FL, USA
- Division of Endocrinology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Christel Hendrieckx
- School of Psychology, Deakin University, Geelong, VIC, Australia
- The Australian Centre for Behavioural Research in Diabetes, Diabetes Victoria, Carlton, VIC, Australia
- Institute for Health Transformation, Deakin University, Geelong, VIC, Australia
| | - Emile Hendriks
- Department of Paediatrics, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | - Richard I G Holt
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health and Care Research Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | | | - Heba M Ismail
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Laura M Jacobsen
- Division of Endocrinology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Suzanne B Johnson
- Department of Behavioral Sciences and Social Medicine, Florida State University College of Medicine, Tallahassee, FL, USA
| | - Leslie E Kolb
- Association of Diabetes Care & Education Specialists, Chicago, IL, USA
| | | | - Karin Lange
- Medical Psychology, Hannover Medical School, Hannover, Germany
| | | | - Åke Lernmark
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Ingrid Libman
- Division of Pediatric Endocrinology and Diabetes, University of Pittsburgh, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Markus Lundgren
- Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Pediatrics, Kristianstad Hospital, Kristianstad, Sweden
| | - David M Maahs
- Division of Endocrinology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - M Loredana Marcovecchio
- Department of Pediatrics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Chantal Mathieu
- Department of Endocrinology, UZ Gasthuisberg, KU Leuven, Leuven, Belgium
| | | | - Holly K O'Donnell
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Tal Oron
- Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shivajirao P Patil
- Department of Family Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Rodica Pop-Busui
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Marian J Rewers
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Desmond A Schatz
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Rifka Schulman-Rosenbaum
- Division of Endocrinology, Long Island Jewish Medical Center, Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, NY, USA
| | - Kimber M Simmons
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Emily K Sims
- Division of Pediatric Endocrinology and Diabetology, Herman B Wells Center for Pediatric Research, Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jay S Skyler
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Laura B Smith
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Cate Speake
- Center for Interventional Immunology and Diabetes Program, Benaroya Research Institute, Seattle, WA, USA
| | - Andrea K Steck
- Department of Pediatrics, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Ksenia N Tonyushkina
- Division of Endocrinology and Diabetes, Baystate Children's Hospital and University of Massachusetts Chan Medical School - Baystate, Springfield, MA, USA
| | - Riitta Veijola
- Research Unit of Clinical Medicine, Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - John M Wentworth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Diane K Wherrett
- Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Jamie R Wood
- Department of Pediatric Endocrinology, Rainbow Babies and Children's Hospital, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Linda A DiMeglio
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
5
|
Iqbal S, Jayyab AA, Alrashdi AM, Shujauddin S, Clua-Espuny JL, Reverté-Villarroya S. The Predictive Potential of C-Peptide in Differentiating Type 1 Diabetes From Type 2 Diabetes in an Outpatient Population in Abu Dhabi. Clin Ther 2024; 46:696-701. [PMID: 39117487 DOI: 10.1016/j.clinthera.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 08/10/2024]
Abstract
PURPOSE We aimed to investigate the predictive potential of plasma connecting peptide (C-peptide) in differentiating type 1 diabetes (T1D) from type 2 diabetes (T2D) and to inform evidence-based diabetes classification criteria. METHODS A retrospective review was performed of all the patients with diabetes visiting an outpatient diabetology, endocrinology, general practice and family medicine tertiary health care center between January 2016 and December 2021. FINDINGS Two hundred twelve individuals with diabetes were included, 85 (44.8%) with T1D and 127 (55.2%) with T2D. Mean (SD) age at diagnosis was 35.9 (15.1) years, and 112 (52.8%) men. Median (interquartile range [IQR]) duration of diabetes was 3.8 (3.0-4.5) years (T1D, 3.9 [3.5-4.6]; T2D, 3.4 [2.4-4.4]; P = 0.001). Body mass index was <18.5 kg/m2 in 5 (2.5%) individuals (T1D, 5; T2D, none), 18.5 to <25 kg/m2 in 57 (28.5%) (T1D, 32; T2D, 25), 25 to <30 kg/m2 in 58 (29%) (T1D, 28; T2D, 30), and >30 kg/m2 in 80 (40.0%) (T1D, 20; T2D, 60). Median (IQR) glycosylated hemoglobin was 7.4% (6.7%-8.5%) (T1D, 8.3% [7.2%-9.9%]; T2D, 7% [6.3%-7.6%]; P = 0.0001). Median (IQR) C-peptide concentration was 0.59 nmol/L (0.01-1.14 nmol/L) (T1D, 0.01 nmol/L [0.003-0.05 nmol/L]; T2D, 1.03 nmol/L [0.70-1.44 nmol/L]; P = 0.0001). C-peptide concentration of ≤0.16 nmol/L showed 92.9% sensitivity, 1-specificity of 2.4%, and AUC of 97.2% (CI, 94.7%-99.6%; P = 0.0001) in differentiating T1D from T2D. IMPLICATIONS To our knowledge, this is the first study in the Middle East and North Africa region highlighting the role of C-peptide in diabetes classification. The estimated cutoff point for C-peptide concentration (≤0.16 nmol/L) will certainly help in accurately classifying the T1D and will rule out the routine clinical judgmental approaches in the region, especially in those scenarios and periods where it is always difficult to diagnose the diabetes type. Quantifying the cutoff for C-peptide is among the vital strengths of this study that will provide a better treatment plan in diabetes care management. Also, we evaluated concomitant glucose levels to rule out the phenomenon of falsely low C-peptide values in the setting of hypoglycemia or severe glucose toxicity. Based on our findings, C-peptide testing could be included in postulating an evidence-based guideline that differentiates T1D from T2D. Despite this, our study has some limitations, including the selection bias due to the retrospective design and low C-peptide levels could be indicative of low pancreatic reserves due to other causes or long-standing T2D, and quantifying these reasons requires additional resources and time.
Collapse
Affiliation(s)
- Sajid Iqbal
- Nursing Department, Universitat Rovira i Virgili, Campus Terres de l'Ebre, Tortosa, Tarragona, Spain; Faculty of Health and Medical Science, Liwa College of Technology, Abu Dhabi, United Arab Emirates.
| | - Abdulrahim Abu Jayyab
- Faculty of Health and Medical Science, Liwa College of Technology, Abu Dhabi, United Arab Emirates
| | - Ayah Mohammad Alrashdi
- Faculty of Health and Medical Science, Liwa College of Technology, Abu Dhabi, United Arab Emirates; Burjeel Hospital, Abu Dhabi, United Arab Emirates
| | | | - Josep Lluis Clua-Espuny
- Primary Health-Care Center EAP Tortosa Est, Institut Català de la Salut, CAP El Temple Plaça Carrilet, Tortosa, Spain; Research Support Unit Terres de l'Ebre, Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAPJGol) (Barcelona), Ebrictus Research Group, Terres de l'Ebre, Tortosa, Spain
| | - Silvia Reverté-Villarroya
- Nursing Department, Universitat Rovira i Virgili, Campus Terres de l'Ebre, Tortosa, Tarragona, Spain; Hospital de Tortosa Verge de la Cinta, Catalan Institute of Health, Pere Virgili Institute, Carretera Esplanetes, Tortosa, Tarragona, Spain
| |
Collapse
|
6
|
Yau C, Danska JS. Cracking the type 1 diabetes code: Genes, microbes, immunity, and the early life environment. Immunol Rev 2024; 325:23-45. [PMID: 39166298 DOI: 10.1111/imr.13362] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Type 1 diabetes (T1D) results from a complex interplay of genetic predisposition, immunological dysregulation, and environmental triggers, that culminate in the destruction of insulin-secreting pancreatic β cells. This review provides a comprehensive examination of the multiple factors underpinning T1D pathogenesis, to elucidate key mechanisms and potential therapeutic targets. Beginning with an exploration of genetic risk factors, we dissect the roles of human leukocyte antigen (HLA) haplotypes and non-HLA gene variants associated with T1D susceptibility. Mechanistic insights gleaned from the NOD mouse model provide valuable parallels to the human disease, particularly immunological intricacies underlying β cell-directed autoimmunity. Immunological drivers of T1D pathogenesis are examined, highlighting the pivotal contributions of both effector and regulatory T cells and the multiple functions of B cells and autoantibodies in β-cell destruction. Furthermore, the impact of environmental risk factors, notably modulation of host immune development by the intestinal microbiome, is examined. Lastly, the review probes human longitudinal studies, unveiling the dynamic interplay between mucosal immunity, systemic antimicrobial antibody responses, and the trajectories of T1D development. Insights garnered from these interconnected factors pave the way for targeted interventions and the identification of biomarkers to enhance T1D management and prevention strategies.
Collapse
Affiliation(s)
- Christopher Yau
- Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Jayne S Danska
- Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Joglekar MV, Kaur S, Pociot F, Hardikar AA. Prediction of progression to type 1 diabetes with dynamic biomarkers and risk scores. Lancet Diabetes Endocrinol 2024; 12:483-492. [PMID: 38797187 DOI: 10.1016/s2213-8587(24)00103-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 05/29/2024]
Abstract
Identifying biomarkers of functional β-cell loss is an important step in the risk stratification of type 1 diabetes. Genetic risk scores (GRS), generated by profiling an array of single nucleotide polymorphisms, are a widely used type 1 diabetes risk-prediction tool. Type 1 diabetes screening studies have relied on a combination of biochemical (autoantibody) and GRS screening methodologies for identifying individuals at high-risk of type 1 diabetes. A limitation of these screening tools is that the presence of autoantibodies marks the initiation of β-cell loss, and is therefore not the best biomarker of progression to early-stage type 1 diabetes. GRS, on the other hand, represents a static biomarker offering a single risk score over an individual's lifetime. In this Personal View, we explore the challenges and opportunities of static and dynamic biomarkers in the prediction of progression to type 1 diabetes. We discuss future directions wherein newer dynamic risk scores could be used to predict type 1 diabetes risk, assess the efficacy of new and emerging drugs to retard, or prevent type 1 diabetes, and possibly replace or further enhance the predictive ability offered by static biomarkers, such as GRS.
Collapse
Affiliation(s)
- Mugdha V Joglekar
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | | | - Flemming Pociot
- Steno Diabetes Center Copenhagen, Herlev, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| | | |
Collapse
|
8
|
Drotar DM, Mojica-Avila AK, Bloss DT, Cohrs CM, Manson CT, Posgai AL, Williams MD, Brusko MA, Phelps EA, Wasserfall CH, Speier S, Atkinson MA. Impaired islet function and normal exocrine enzyme secretion occur with low inter-regional variation in type 1 diabetes. Cell Rep 2024; 43:114346. [PMID: 38850534 PMCID: PMC11251461 DOI: 10.1016/j.celrep.2024.114346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/03/2024] [Accepted: 05/24/2024] [Indexed: 06/10/2024] Open
Abstract
Histopathological heterogeneity in the human pancreas is well documented; however, functional evidence at the tissue level is scarce. Herein, we investigate in situ glucose-stimulated islet and carbachol-stimulated acinar cell secretion across the pancreas head (PH), body (PB), and tail (PT) regions in donors without diabetes (ND; n = 15), positive for one islet autoantibody (1AAb+; n = 7), and with type 1 diabetes (T1D; <14 months duration, n = 5). Insulin, glucagon, pancreatic amylase, lipase, and trypsinogen secretion along with 3D tissue morphometrical features are comparable across regions in ND. In T1D, insulin secretion and beta-cell volume are significantly reduced within all regions, while glucagon and enzymes are unaltered. Beta-cell volume is lower despite normal insulin secretion in 1AAb+, resulting in increased volume-adjusted insulin secretion versus ND. Islet and acinar cell secretion in 1AAb+ are consistent across the PH, PB, and PT. This study supports low inter-regional variation in pancreas slice function and, potentially, increased metabolic demand in 1AAb+.
Collapse
Affiliation(s)
- Denise M Drotar
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | - Ana Karen Mojica-Avila
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), München, Neuherberg, Germany
| | - Drew T Bloss
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | - Christian M Cohrs
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), München, Neuherberg, Germany
| | - Cameron T Manson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA; J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | - MacKenzie D Williams
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | - Maigan A Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA
| | - Edward A Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Clive H Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA; Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Stephan Speier
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany; Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), München, Neuherberg, Germany
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL 32610, USA; Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA.
| |
Collapse
|
9
|
Melchiorre CK, Lynes MD, Bhandari S, Su SC, Potts CM, Thees AV, Norris CE, Liaw L, Tseng YH, Lynes MA. Extracellular metallothionein as a therapeutic target in the early progression of type 1 diabetes. Cell Stress Chaperones 2024; 29:312-325. [PMID: 38490439 PMCID: PMC10990868 DOI: 10.1016/j.cstres.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/23/2024] [Accepted: 03/11/2024] [Indexed: 03/17/2024] Open
Abstract
Type 1 diabetes (T1D) is characterized by lymphocyte infiltration into the pancreatic islets of Langerhans, leading to the destruction of insulin-producing beta cells and uncontrolled hyperglycemia. In the nonobese diabetic (NOD) murine model of T1D, the onset of this infiltration starts several weeks before glucose dysregulation and overt diabetes. Recruitment of immune cells to the islets is mediated by several chemotactic cytokines, including CXCL10, while other cytokines, including SDF-1α, can confer protective effects. Global gene expression studies of the pancreas from prediabetic NOD mice and single-cell sequence analysis of human islets from prediabetic, autoantibody-positive patients showed an increased expression of metallothionein (MT), a small molecular weight, cysteine-rich metal-binding stress response protein. We have shown that beta cells can release MT into the extracellular environment, which can subsequently enhance the chemotactic response of Th1 cells to CXCL10 and interfere with the chemotactic response of Th2 cells to SDF-1α. These effects can be blocked in vitro with a monoclonal anti-MT antibody, clone UC1MT. When administered to NOD mice before the onset of diabetes, UC1MT significantly reduces the development of T1D. Manipulation of extracellular MT may be an important approach to preserving beta cell function and preventing the development of T1D.
Collapse
Affiliation(s)
- Clare K Melchiorre
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Matthew D Lynes
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
| | - Sadikshya Bhandari
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Sheng-Chiang Su
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA; Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Christian M Potts
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
| | - Amy V Thees
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Carol E Norris
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Lucy Liaw
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Michael A Lynes
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
10
|
Wang J, Wan K, Chang X, Mao RF. Association of autoimmune thyroid disease with type 1 diabetes mellitus and its ultrasonic diagnosis and management. World J Diabetes 2024; 15:348-360. [PMID: 38591076 PMCID: PMC10999045 DOI: 10.4239/wjd.v15.i3.348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/14/2023] [Accepted: 01/18/2024] [Indexed: 03/15/2024] Open
Abstract
As a common hyperglycemic disease, type 1 diabetes mellitus (T1DM) is a complicated disorder that requires a lifelong insulin supply due to the immune-mediated destruction of pancreatic β cells. Although it is an organ-specific autoimmune disorder, T1DM is often associated with multiple other autoimmune disorders. The most prevalent concomitant autoimmune disorder occurring in T1DM is autoimmune thyroid disease (AITD), which mainly exhibits two extremes of phenotypes: hyperthyroidism [Graves' disease (GD)] and hypo-thyroidism [Hashimoto's thyroiditis, (HT)]. However, the presence of comorbid AITD may negatively affect metabolic management in T1DM patients and thereby may increase the risk for potential diabetes-related complications. Thus, routine screening of thyroid function has been recommended when T1DM is diagnosed. Here, first, we summarize current knowledge regarding the etiology and pathogenesis mechanisms of both diseases. Subsequently, an updated review of the association between T1DM and AITD is offered. Finally, we provide a relatively detailed review focusing on the application of thyroid ultrasonography in diagnosing and managing HT and GD, suggesting its critical role in the timely and accurate diagnosis of AITD in T1DM.
Collapse
Affiliation(s)
- Jin Wang
- Department of Ultrasound Medicine, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing 211200, Jiangsu Province, China
| | - Ke Wan
- Faculty of Medicine and Health, The University of Sydney, Camperdown NSW 2050, Australia
| | - Xin Chang
- Department of Ultrasound Medicine, Nanjing Lishui People’s Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing 211200, Jiangsu Province, China
| | - Rui-Feng Mao
- School of Life Science, Huaiyin Normal University, Huai'an 223300, Jiangsu Province, China
| |
Collapse
|
11
|
Merolla A, De Lorenzo R, Ferrannini G, Renzi C, Ulivi F, Bazzigaluppi E, Lampasona V, Bosi E. Universal screening for early detection of chronic autoimmune, metabolic and cardiovascular diseases in the general population using capillary blood (UNISCREEN): low-risk interventional, single-centre, pilot study protocol. BMJ Open 2024; 14:e078983. [PMID: 38448070 PMCID: PMC10916121 DOI: 10.1136/bmjopen-2023-078983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
INTRODUCTION Chronic autoimmune (type 1 diabetes and coeliac disease) and metabolic/cardiovascular (type 2 diabetes, dyslipidaemia, hypertension) diseases are highly prevalent across all age ranges representing a major public health burden. Universal screening for prediction/early identification of these conditions is a potential tool for reducing their impact on the general population. The aim of this study is to assess whether universal screening using capillary blood sampling is feasible at a population-based level. METHODS AND ANALYSIS This is a low-risk interventional, single-centre, pilot study for a population-based screening programme denominated UNISCREEN. Participants are volunteers aged 1-100 who reside in the town of Cantalupo (Milan, Italy) undergoing: (1) interview collecting demographics, anthropometrics and medical history; (2) capillary blood collection for measurement of type 1 diabetes and coeliac disease-specific autoantibodies and immediate measurement of glucose, glycated haemoglobin and lipid panel by point-of-care devices; (3) venous blood sampling to confirm autoantibody-positivity; (4) blood pressure measurement; (5) fulfilment of a feasibility and acceptability questionnaire. The outcomes are the assessment of feasibility and acceptability of capillary blood screening, the prevalence of presymptomatic type 1 diabetes and undiagnosed coeliac disease, distribution of glucose categories, lipid panel and estimate of cardiovascular risk in the study population. With approximately 3000 inhabitants, the screened population is expected to encompass at least half of its size, approaching nearly 1500 individuals. ETHICS AND DISSEMINATION This protocol and the informed consent forms have been reviewed and approved by the San Raffaele Hospital Ethics Committee (approval number: 131/INT/2022). Written informed consent is obtained from all study participants or their parents if aged <18. Results will be published in scientific journals and presented at meetings. CONCLUSIONS If proven feasible and acceptable, this universal screening model would pave the way for larger-scale programmes, providing an opportunity for the implementation of innovative public health programmes in the general population. TRIAL REGISTRATION NUMBER NCT05841719.
Collapse
Affiliation(s)
| | | | | | - Cristina Renzi
- Vita-Salute San Raffaele University, Milano, Italy
- Behavioural Science and Health, Institute of Epidemiology & Health Care, University College London, London, UK
| | | | | | - Vito Lampasona
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - Emanuele Bosi
- Vita-Salute San Raffaele University, Milano, Italy
- Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
12
|
Drotar DM, Mojica-Avila AK, Bloss DT, Cohrs CM, Manson CT, Posgai AL, Williams MD, Brusko MA, Phelps EA, Wasserfall CH, Speier S, Atkinson MA. Impaired islet function with normal exocrine enzyme secretion is consistent across the head, body, and tail pancreas regions in type 1 diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.08.579175. [PMID: 38405840 PMCID: PMC10888906 DOI: 10.1101/2024.02.08.579175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Histopathological heterogeneity in human pancreas has been well documented; however, functional evidence at the tissue level is scarce. Herein we investigated in situ glucose-stimulated islet and carbachol-stimulated acinar cell secretion across the pancreas head (PH), body (PB), and tail (PT) regions in no diabetes (ND, n=15), single islet autoantibody-positive (1AAb+, n=7), and type 1 diabetes donors (T1D, <14 months duration, n=5). Insulin, glucagon, pancreatic amylase, lipase, and trypsinogen secretion along with 3D tissue morphometrical features were comparable across the regions in ND. In T1D, insulin secretion and beta-cell volume were significantly reduced within all regions, while glucagon and enzymes were unaltered. Beta-cell volume was lower despite normal insulin secretion in 1AAb+, resulting in increased volume-adjusted insulin secretion versus ND. Islet and acinar cell secretion in 1AAb+ were consistent across PH, PB and PT. This study supports low inter-regional variation in pancreas slice function and potentially, increased metabolic demand in 1AAb+.
Collapse
Affiliation(s)
- Denise M. Drotar
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
| | - Ana Karen Mojica-Avila
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Drew T. Bloss
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
| | - Christian M. Cohrs
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Cameron T. Manson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL USA
| | - Amanda L. Posgai
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
| | - MacKenzie D. Williams
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
| | - Maigan A. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
| | - Edward A. Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL USA
| | - Clive H. Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL USA
| | - Stephan Speier
- Institute of Physiology, Faculty of Medicine, Technische Universität Dresden, Germany
- Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Clinic Carl Gustav Carus of Technische Universität Dresden, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Mark A. Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL USA
| |
Collapse
|
13
|
Alcazar O, Chuang ST, Ren G, Ogihara M, Webb-Robertson BJM, Nakayasu ES, Buchwald P, Abdulreda MH. A Composite Biomarker Signature of Type 1 Diabetes Risk Identified via Augmentation of Parallel Multi-Omics Data from a Small Cohort. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.09.579673. [PMID: 38405796 PMCID: PMC10888829 DOI: 10.1101/2024.02.09.579673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Background Biomarkers of early pathogenesis of type 1 diabetes (T1D) are crucial to enable effective prevention measures in at-risk populations before significant damage occurs to their insulin producing beta-cell mass. We recently introduced the concept of integrated parallel multi-omics and employed a novel data augmentation approach which identified promising candidate biomarkers from a small cohort of high-risk T1D subjects. We now validate selected biomarkers to generate a potential composite signature of T1D risk. Methods Twelve candidate biomarkers, which were identified in the augmented data and selected based on their fold-change relative to healthy controls and cross-reference to proteomics data previously obtained in the expansive TEDDY and DAISY cohorts, were measured in the original samples by ELISA. Results All 12 biomarkers had established connections with lipid/lipoprotein metabolism, immune function, inflammation, and diabetes, but only 7 were found to be markedly changed in the high-risk subjects compared to the healthy controls: ApoC1 and PON1 were reduced while CETP, CD36, FGFR1, IGHM, PCSK9, SOD1, and VCAM1 were elevated. Conclusions Results further highlight the promise of our data augmentation approach in unmasking important patterns and pathologically significant features in parallel multi-omics datasets obtained from small sample cohorts to facilitate the identification of promising candidate T1D biomarkers for downstream validation. They also support the potential utility of a composite biomarker signature of T1D risk characterized by the changes in the above markers.
Collapse
|
14
|
Besser REJ, Long AE, Owen KR, Law R, Birks JS, Pearce O, Williams CL, Scudder CL, McDonald TJ, Todd JA. Transdermal Blood Sampling for C-Peptide Is a Minimally Invasive, Reliable Alternative to Venous Sampling in Children and Adults With Type 1 Diabetes. Diabetes Care 2024; 47:239-245. [PMID: 38087932 DOI: 10.2337/dc23-1379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/02/2023] [Indexed: 01/21/2024]
Abstract
OBJECTIVE C-peptide and islet autoantibodies are key type 1 diabetes biomarkers, typically requiring venous sampling, which limits their utility. We assessed transdermal capillary blood (TCB) collection as a practical alternative. RESEARCH DESIGN AND METHODS Ninety-one individuals (71 with type 1 diabetes, 20 control; individuals with type 1 diabetes: aged median 14.8 years [interquartile range (IQR) 9.1-17.1], diabetes duration 4.0 years [1.5-7.7]; control individuals: 42.2 years [38.0-52.1]) underwent contemporaneous venous and TCB sampling for measurement of plasma C-peptide. Participants with type 1 diabetes also provided venous serum and plasma, and TCB plasma for measurement of autoantibodies to glutamate decarboxylase, islet antigen-2, and zinc transporter 8. The ability of TCB plasma to detect significant endogenous insulin secretion (venous C-peptide ≥200 pmol/L) was compared along with agreement in levels, using Bland-Altman. Venous serum was compared with venous and TCB plasma for detection of autoantibodies, using established thresholds. Acceptability was assessed by age-appropriate questionnaire. RESULTS Transdermal sampling took a mean of 2.35 min (SD 1.49). Median sample volume was 50 µL (IQR 40-50) with 3 of 91 (3.3%) failures, and 13 of 88 (14.7%) <35 µL. TCB C-peptide showed good agreement with venous plasma (mean venous ln[C-peptide] - TCB ln[C-peptide] = 0.008, 95% CI [-0.23, 0.29], with 100% [36 of 36] sensitivity/100% [50 of 50] specificity to detect venous C-peptide ≥200 pmol/L). Where venous serum in multiple autoantibody positive TCB plasma agreed in 22 of 32 (sensitivity 69%), comparative specificity was 35 of 36 (97%). TCB was preferred to venous sampling (type 1 diabetes: 63% vs. 7%; 30% undecided). CONCLUSIONS Transdermal capillary testing for C-peptide is a sensitive, specific, and acceptable alternative to venous sampling; TCB sampling for islet autoantibodies needs further assessment.
Collapse
Affiliation(s)
- Rachel E J Besser
- Juvenile Diabetes Research Foundation/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, Oxford National Institute for Health Research (NIHR) Biomedical Research Centre, University of Oxford, Oxford, U.K
- Department of Paediatric Diabetes, Oxford Children's Hospital, John Radcliffe Hospital, Oxford, U.K
| | - Anna E Long
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Katharine R Owen
- Oxford NIHR Biomedical Research Centre, University of Oxford, Oxford, U.K
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
| | - Rebecca Law
- Department of Paediatric Diabetes, Oxford Children's Hospital, John Radcliffe Hospital, Oxford, U.K
| | - Jacqueline S Birks
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Centre for Statistics in Medicine, University of Oxford, Oxford, U.K
| | - Olivia Pearce
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Claire L Williams
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Claire L Scudder
- Juvenile Diabetes Research Foundation/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, Oxford National Institute for Health Research (NIHR) Biomedical Research Centre, University of Oxford, Oxford, U.K
| | - Timothy J McDonald
- Academic Department of Blood Sciences, Royal Devon University Hospital, Exeter, U.K
- Exeter NIHR Biomedical Research Centre, University of Exeter, Exeter, U.K
| | - John A Todd
- Juvenile Diabetes Research Foundation/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, Oxford National Institute for Health Research (NIHR) Biomedical Research Centre, University of Oxford, Oxford, U.K
| |
Collapse
|
15
|
Tatum JD, Hornung L, Bellin MD, Elder DA, Thompson T, Vitale DS, Wasserfall CH, Shah AS, Abu-El-Haija M. High Rate of Islets Autoimmunity in Pediatric Patients with Index Admission of Acute Pancreatitis. Pediatr Diabetes 2023; 2023:9170497. [PMID: 39600796 PMCID: PMC11594539 DOI: 10.1155/2023/9170497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
Introduction The underlying pathophysiology of diabetes mellitus after acute pancreatitis is unknown and overall risk of developing diabetes postacute pancreatitis in children is understudied. The objective of our study was to describe the frequency of islet cell autoimmunity and abnormal glucose testing in pediatric patients in the year following their index case of acute pancreatitis. Materials and Methods Data were obtained from a single-center observational cohort study of patients with their first episode of acute pancreatitis. Islet cell autoantibody titers were measured on stored plasma collected from acute pancreatitis diagnosis, at 3 months and at 12 months postacute pancreatitis attack. Abnormal glucose testing was defined as the presence of prediabetes or diabetes, as defined by American Diabetes Association criteria. Results Eighty-four patients with acute pancreatitis and islet cell autoantibody data were included, 71 had available glucose measures. Median age at first acute pancreatitis attack was 14 years (IQR 8.7-16.3) and 45/84 (54%) were females. Twenty-four patients (29%) were positive for at least one of four islet cell autoantibodies (IAA, GADA, IA-2A, and ZnT8A) and 6 (7%) had two or more positive islet cell autoantibodies. Nineteen patients out of 71 (27%) had abnormal glucose testing at or postacute pancreatitis diagnosis. A higher proportion (37%, 7/19) with abnormal glucose testing had severe acute pancreatitis compared to those with normal glucose testing (13%, 7/52) (p = 0:04). Patients with normal glucose testing were more likely to be positive for one or more islet cell autoantibodies (31%, 16/52) compared to those with abnormal glucose testing (0%, 0/19) (p = 0:004). Conclusions Islet cell autoimmunity is more common in children after their index acute pancreatitis attack (29%) than in the general population (7%-8%). While the frequency of prediabetes and diabetes postacute pancreatitis is high, other mechanisms besides islet cell autoimmunity are responsible.
Collapse
Affiliation(s)
- Jonathan D. Tatum
- Division of Pediatric Endocrinology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Lindsey Hornung
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center Cincinnati, Cincinnati, USA
| | - Melena D. Bellin
- Division of Pediatric Endocrinology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
- Department of Surgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Deborah A. Elder
- Division of Pediatric Endocrinology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Tyler Thompson
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - David S. Vitale
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Clive H. Wasserfall
- Department of Pathology, Immunology, and Laboratory Science, University of Florida, Gainesville, USA
- College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Amy S. Shah
- Division of Pediatric Endocrinology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Maisam Abu-El-Haija
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
16
|
Mameli C, Triolo TM, Chiarelli F, Rewers M, Zuccotti G, Simmons KM. Lessons and Gaps in the Prediction and Prevention of Type 1 Diabetes. Pharmacol Res 2023; 193:106792. [PMID: 37201589 DOI: 10.1016/j.phrs.2023.106792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/01/2023] [Accepted: 05/08/2023] [Indexed: 05/20/2023]
Abstract
Type 1 diabetes (T1D) is a serious chronic autoimmune condition. Even though the root cause of T1D development has yet to be determined, enough is known about the natural history of T1D pathogenesis to allow study of interventions that may delay or even prevent the onset of hyperglycemia and clinical T1D. Primary prevention aims to prevent the onset of beta cell autoimmunity in asymptomatic people at high genetic risk for T1D. Secondary prevention strategies aim to preserve functional beta cells once autoimmunity is present, and tertiary prevention aims to initiate and extend partial remission of beta cell destruction after the clinical onset of T1D. The approval of teplizumab in the United States to delay the onset of clinical T1D marks an impressive milestone in diabetes care. This treatment opens the door to a paradigm shift in T1D care. People with T1D risk need to be identified early by measuring T1D related islet autoantibodies. Identifying people with T1D before they have symptoms will facilitate better understanding of pre-symptomatic T1D progression and T1D prevention strategies that may be effective.
Collapse
Affiliation(s)
- Chiara Mameli
- Department of Pediatrics, V. Buzzi Children's Hospital, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy.
| | - Taylor M Triolo
- Barbara Davis Center for Diabetes, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045
| | | | - Marian Rewers
- Barbara Davis Center for Diabetes, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045
| | - Gianvincenzo Zuccotti
- Department of Pediatrics, V. Buzzi Children's Hospital, Milan, Italy; Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Kimber M Simmons
- Barbara Davis Center for Diabetes, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045
| |
Collapse
|
17
|
Ochocińska A, Wysocka-Mincewicz M, Świderska J, Cukrowska B. Selected Serum Markers Associated with Pathogenesis and Clinical Course of Type 1 Diabetes in Pediatric Patients-The Effect of Disease Duration. J Clin Med 2023; 12:2151. [PMID: 36983153 PMCID: PMC10051659 DOI: 10.3390/jcm12062151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Biochemical abnormalities in the course of type 1 diabetes (T1D) may cause the production/activation of various proteins and peptides influencing treatment and causing a risk of complications. The aim of this study was to assess concentrations of selected serum substances involved in the pathogenesis and course of T1D and to correlate their concentrations with the duration of T1D. The study included patients with T1D (n = 156) at the age of 3-17, who were divided according to the duration of the disease into those newly diagnosed (n = 30), diagnosed after 3-5 (n = 77), 6-7 (n = 25), and over 7 (n = 24) years from the onset of T1D, and age-matched healthy controls (n = 30). Concentrations of amylin (IAPP), proamylin (proIAPP), catestatin (CST), chromogranin A (ChgA), nerve growth factor (NFG), platelet-activating factor (PAF), uromodulin (UMOD), and intestinal fatty acid binding protein (I-FABP) were measured in sera using immunoenzymatic tests. There were significant differences in concentrations of all the substances except UMOD and NGF between T1D patients and healthy children. The duration of the disease affected concentrations of CST, ChgA, PAF, and NGF, i.e., proteins/peptides which could have an impact on the course of T1D and the development of complications. In long-term patients, a decrease in concentrations of CST and ChgA, and an increase in PAF concentrations were found. In the case of NGF, a decrease was observed after the initial high values, followed by an increase over 7 years after T1D diagnosis. Concluding, the results show that concentrations of selected serum indicators may change in the course of T1D. Further studies are needed to establish whether these indicators could be used in the context of predicting long-term complications.
Collapse
Affiliation(s)
- Agnieszka Ochocińska
- Department of Biochemistry, Radioimmunology and Experimental Medicine, The Children’s Memorial Health Institute, Aleja Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Marta Wysocka-Mincewicz
- Clinic of Endocrinology and Diabetology, The Children’s Memorial Health Institute, Aleja Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Jolanta Świderska
- Clinic of Endocrinology and Diabetology, The Children’s Memorial Health Institute, Aleja Dzieci Polskich 20, 04-730 Warsaw, Poland
| | - Bożena Cukrowska
- Department of Pathomorphology, The Children’s Memorial Health Institute, Aleja Dzieci Polskich 20, 04-730 Warsaw, Poland
| |
Collapse
|
18
|
Ng K, Anand V, Stavropoulos H, Veijola R, Toppari J, Maziarz M, Lundgren M, Waugh K, Frohnert BI, Martin F, Lou O, Hagopian W, Achenbach P. Quantifying the utility of islet autoantibody levels in the prediction of type 1 diabetes in children. Diabetologia 2023; 66:93-104. [PMID: 36195673 PMCID: PMC9729160 DOI: 10.1007/s00125-022-05799-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS The aim of this study was to explore the utility of islet autoantibody (IAb) levels for the prediction of type 1 diabetes in autoantibody-positive children. METHODS Prospective cohort studies in Finland, Germany, Sweden and the USA followed 24,662 children at increased genetic or familial risk of developing islet autoimmunity and diabetes. For the 1403 who developed IAbs (523 of whom developed diabetes), levels of autoantibodies against insulin (IAA), glutamic acid decarboxylase (GADA) and insulinoma-associated antigen-2 (IA-2A) were harmonised for analysis. Diabetes prediction models using multivariate logistic regression with inverse probability censored weighting (IPCW) were trained using 10-fold cross-validation. Discriminative power for disease was estimated using the IPCW concordance index (C index) with 95% CI estimated via bootstrap. RESULTS A baseline model with covariates for data source, sex, diabetes family history, HLA risk group and age at seroconversion with a 10-year follow-up period yielded a C index of 0.61 (95% CI 0.58, 0.63). The performance improved after adding the IAb positivity status for IAA, GADA and IA-2A at seroconversion: C index 0.72 (95% CI 0.71, 0.74). Using the IAb levels instead of positivity indicators resulted in even better performance: C index 0.76 (95% CI 0.74, 0.77). The predictive power was maintained when using the IAb levels alone: C index 0.76 (95% CI 0.75, 0.76). The prediction was better for shorter follow-up periods, with a C index of 0.82 (95% CI 0.81, 0.83) at 2 years, and remained reasonable for longer follow-up periods, with a C index of 0.76 (95% CI 0.75, 0.76) at 11 years. Inclusion of the results of a third IAb test added to the predictive power, and a suitable interval between seroconversion and the third test was approximately 1.5 years, with a C index of 0.78 (95% CI 0.77, 0.78) at 10 years follow-up. CONCLUSIONS/INTERPRETATION Consideration of quantitative patterns of IAb levels improved the predictive power for type 1 diabetes in IAb-positive children beyond qualitative IAb positivity status.
Collapse
Affiliation(s)
| | | | | | - Riitta Veijola
- Department of Pediatrics, PEDEGO Research Unit, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Jorma Toppari
- Institute of Biomedicine and Centre for Population Health Research, University of Turku, Turku, Finland
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Marlena Maziarz
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Markus Lundgren
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
- Department of Pediatrics, Kristianstad Hospital, Kristianstad, Sweden
| | - Kathy Waugh
- Barbara Davis Center for Diabetes, University of Colorado, Denver, CO, USA
| | | | | | | | | | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany.
| |
Collapse
|
19
|
Strollo R, Vinci C, Man YKS, Bruzzaniti S, Piemonte E, Alhamar G, Briganti SI, Malandrucco I, Tramontana F, Fanali C, Garnett J, Buccafusca R, Guyer P, Mamula M, James EA, Pozzilli P, Ludvigsson J, Winyard PG, Galgani M, Nissim A. Autoantibody and T cell responses to oxidative post-translationally modified insulin neoantigenic peptides in type 1 diabetes. Diabetologia 2023; 66:132-146. [PMID: 36207582 PMCID: PMC9729141 DOI: 10.1007/s00125-022-05812-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/28/2022] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS Antibodies specific to oxidative post-translational modifications (oxPTM) of insulin (oxPTM-INS) are present in most individuals with type 1 diabetes, even before the clinical onset. However, the antigenic determinants of such response are still unknown. In this study, we investigated the antibody response to oxPTM-INS neoepitope peptides (oxPTM-INSPs) and evaluated their ability to stimulate humoral and T cell responses in type 1 diabetes. We also assessed the concordance between antibody and T cell responses to the oxPTM-INS neoantigenic peptides. METHODS oxPTM-INS was generated by exposing insulin to various reactive oxidants. The insulin fragments resulting from oxPTM were fractionated by size-exclusion chromatography further to ELISA and LC-MS/MS analysis to identify the oxidised peptide neoepitopes. Immunogenic peptide candidates were produced and then modified in house or designed to incorporate in silico-oxidised amino acids during synthesis. Autoantibodies to the oxPTM-INSPs were tested by ELISA using sera from 63 participants with new-onset type 1 diabetes and 30 control participants. An additional 18 fresh blood samples from participants with recently diagnosed type 1 diabetes, five with established disease, and from 11 control participants were used to evaluate, in parallel, CD4+ and CD8+ T cell activation by oxPTM-INSPs. RESULTS We observed antibody and T cell responses to three out of six LC-MS/MS-identified insulin peptide candidates: A:12-21 (SLYQLENYCN, native insulin peptide 3 [Nt-INSP-3]), B:11-30 (LVEALYLVCGERGFFYTPKT, Nt-INSP-4) and B:21-30 (ERGFFYTPKT, Nt-INSP-6). For Nt-INSP-4 and Nt-INSP-6, serum antibody binding was stronger in type 1 diabetes compared with healthy control participants (p≤0.02), with oxidised forms of ERGFFYTPKT, oxPTM-INSP-6 conferring the highest antibody binding (83% binders to peptide modified in house by hydroxyl radical [●OH] and >88% to in silico-oxidised peptide; p≤0.001 vs control participants). Nt-INSP-4 induced the strongest T cell stimulation in type 1 diabetes compared with control participants for both CD4+ (p<0.001) and CD8+ (p=0.049). CD4+ response to oxPTM-INSP-6 was also commoner in type 1 diabetes than in control participants (66.7% vs 27.3%; p=0.039). Among individuals with type 1 diabetes, the CD4+ response to oxPTM-INSP-6 was more frequent than to Nt-INSP-6 (66.7% vs 27.8%; p=0.045). Overall, 44.4% of patients showed a concordant autoimmune response to oxPTM-INSP involving simultaneously CD4+ and CD8+ T cells and autoantibodies. CONCLUSIONS/INTERPRETATION Our findings support the concept that oxidative stress, and neoantigenic epitopes of insulin, may be involved in the immunopathogenesis of type 1 diabetes.
Collapse
Affiliation(s)
- Rocky Strollo
- Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Chiara Vinci
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Y K Stella Man
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Sara Bruzzaniti
- Institute for Experimental Endocrinology and Oncology 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
- Department of Biology, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Erica Piemonte
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Ghadeer Alhamar
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Silvia Irina Briganti
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Ilaria Malandrucco
- The UOSD of Endocrinology and Metabolic Diseases, Azienda Sanitaria Locale (ASL) Frosinone, Frosinone, Italy
| | - Flavia Tramontana
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Chiara Fanali
- Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, Rome, Italy
| | - James Garnett
- Centre for Host-Microbiome Interactions, Dental Institute, King's College London, London, UK
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Roberto Buccafusca
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Perrin Guyer
- Program for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Mark Mamula
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Eddie A James
- Program for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Paolo Pozzilli
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Johnny Ludvigsson
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Crown Princess Victoria Children's Hospital, Linköping University, Linköping, Sweden
| | - Paul G Winyard
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, St Luke's Campus, Exeter, UK
| | - Mario Galgani
- Institute for Experimental Endocrinology and Oncology 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Ahuva Nissim
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
20
|
Bendor-Samuel OM, Wishlade T, Willis L, Aley P, Choi E, Craik R, Mujadidi Y, Mounce G, Roseman F, De La Horra Gozalo A, Bland J, Taj N, Smith I, Ziegler AG, Bonifacio E, Winkler C, Haupt F, Todd JA, Servais L, Snape MD, Vatish M. Successful integration of newborn genetic testing into UK routine screening using prospective consent to determine eligibility for clinical trials. Arch Dis Child 2023; 108:26-30. [PMID: 36171064 PMCID: PMC9763160 DOI: 10.1136/archdischild-2022-324270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 09/09/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVE INGR1D (INvestigating Genetic Risk for type 1 Diabetes) was a type 1 diabetes (T1D) genetic screening study established to identify participants for a primary prevention trial (POInT, Primary Oral Insulin Trial). METHODS The majority of participants were recruited by research midwives in antenatal clinics from 18 weeks' gestation. Using the NHS Newborn Bloodspot Screening Programme (NBSP) infrastructure, participants enrolled in INGR1D had an extra sample taken from their day 5 bloodspot card sent for T1D genetic screening. Those at an increased risk of T1D were informed of the result, given education about T1D and the opportunity to take part in POInT. RESULTS Between April 2018 and November 2020, 66% of women approached about INGR1D chose to participate. 15 660 babies were enrolled into INGR1D and 14 731 blood samples were processed. Of the processed samples, 157 (1%) had confirmed positive results, indicating an increased risk of T1D, of whom a third (n=49) enrolled into POInT (20 families were unable to participate in POInT due to COVID-19 lockdown restrictions). CONCLUSION The use of prospective consent to perform personalised genetic testing on samples obtained through the routine NBSP represents a novel mechanism for clinical genetic research in the UK and provides a model for further population-based genetic studies in the newborn.
Collapse
Affiliation(s)
| | - Tabitha Wishlade
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, Oxfordshire, UK
| | - Louise Willis
- Oxford Vaccine Group, University of Oxford, Oxford, Oxfordshire, UK
| | - Parvinder Aley
- Oxford Vaccine Group, University of Oxford, Oxford, Oxfordshire, UK
| | - Edward Choi
- Oxford Vaccine Group, University of Oxford, Oxford, Oxfordshire, UK
| | - Rachel Craik
- Oxford Vaccine Group, University of Oxford, Oxford, Oxfordshire, UK
| | - Yama Mujadidi
- Oxford Vaccine Group, University of Oxford, Oxford, Oxfordshire, UK
| | - Ginny Mounce
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, Oxfordshire, UK
| | - Fenella Roseman
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, Oxfordshire, UK
| | | | - James Bland
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, Oxfordshire, UK
| | - Nazia Taj
- Oxford Screening Laboratory, Department of Clinical Biochemistry, Oxford University Hospitals NHS Foundation Trust, Oxford, Oxfordshire, UK
| | - Ian Smith
- Oxford Screening Laboratory, Department of Clinical Biochemistry, Oxford University Hospitals NHS Foundation Trust, Oxford, Oxfordshire, UK
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany,Technical University Munich, School of Medicine, Forschergruppe Diabetes at Klinikum rechts der Isar, Munich, Germany
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christiane Winkler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Florian Haupt
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - John A Todd
- Wellcome Centre for Human Genetics, University of Oxford Nuffield Department of Medicine, Oxford, Oxfordshire, UK,NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Laurent Servais
- Division of Child Neurology, Centre de Références des Maladies Neuromusculaires, Department of Pediatrics, Université de Liège, Liege, Belgium,MDUK Neuromuscular Centre, University of Oxford Department of Paediatrics, Oxford, Oxfordshire, UK
| | - Matthew D Snape
- Oxford Vaccine Group, University of Oxford, Oxford, Oxfordshire, UK,NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, Oxfordshire, UK .,Wellcome Centre for Human Genetics, University of Oxford Nuffield Department of Medicine, Oxford, Oxfordshire, UK
| | | |
Collapse
|
21
|
Weiss A, Zapardiel-Gonzalo J, Voss F, Jolink M, Stock J, Haupt F, Kick K, Welzhofer T, Heublein A, Winkler C, Achenbach P, Ziegler AG, Bonifacio E. Progression likelihood score identifies substages of presymptomatic type 1 diabetes in childhood public health screening. Diabetologia 2022; 65:2121-2131. [PMID: 36028774 PMCID: PMC9630406 DOI: 10.1007/s00125-022-05780-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/07/2022] [Indexed: 01/11/2023]
Abstract
AIMS/HYPOTHESIS The aim of this study was to develop strategies that identify children from the general population who have late-stage presymptomatic type 1 diabetes and may, therefore, benefit from immune intervention. METHODS We tested children from Bavaria, Germany, aged 1.75-10 years, enrolled in the Fr1da public health screening programme for islet autoantibodies (n=154,462). OGTT and HbA1c were assessed in children with multiple islet autoantibodies for diagnosis of presymptomatic stage 1 (normoglycaemia) or stage 2 (dysglycaemia) type 1 diabetes. Cox proportional hazards and penalised logistic regression of autoantibody, genetic, metabolic and demographic information were used to develop a progression likelihood score to identify children with stage 1 type 1 diabetes who progressed to stage 3 (clinical) type 1 diabetes within 2 years. RESULTS Of 447 children with multiple islet autoantibodies, 364 (81.4%) were staged. Undiagnosed stage 3 type 1 diabetes, presymptomatic stage 2, and stage 1 type 1 diabetes were detected in 41 (0.027% of screened children), 30 (0.019%) and 293 (0.19%) children, respectively. The 2 year risk for progression to stage 3 type 1 diabetes was 48% (95% CI 34, 58) in children with stage 2 type 1 diabetes (annualised risk, 28%). HbA1c, islet antigen-2 autoantibody positivity and titre, and the 90 min OGTT value were predictors of progression in children with stage 1 type 1 diabetes. The derived progression likelihood score identified substages corresponding to ≤90th centile (stage 1a, n=258) and >90th centile (stage 1b, n=29; 0.019%) of stage 1 children with a 4.1% (95% CI 1.4, 6.7) and 46% (95% CI 21, 63) 2 year risk of progressing to stage 3 type 1 diabetes, respectively. CONCLUSIONS/INTERPRETATION Public health screening for islet autoantibodies found 0.027% of children to have undiagnosed clinical type 1 diabetes and 0.038% to have undiagnosed presymptomatic stage 2 or stage 1b type 1 diabetes, with 50% risk to develop clinical type 1 diabetes within 2 years.
Collapse
Affiliation(s)
- Andreas Weiss
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
| | - Jose Zapardiel-Gonzalo
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
| | - Franziska Voss
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Manja Jolink
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Joanna Stock
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Florian Haupt
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
- Forschergruppe Diabetes e.V. at Helmholtz Zentrum München, Munich, Germany
| | - Kerstin Kick
- Technical University Munich, School of Medicine, Forschergruppe Diabetes at Klinikum rechts der Isar, Munich, Germany
| | - Tiziana Welzhofer
- Technical University Munich, School of Medicine, Forschergruppe Diabetes at Klinikum rechts der Isar, Munich, Germany
| | - Anja Heublein
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Christiane Winkler
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
- Forschergruppe Diabetes e.V. at Helmholtz Zentrum München, Munich, Germany
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
- German Center for Diabetes Research (DZD), Munich, Germany
- Forschergruppe Diabetes e.V. at Helmholtz Zentrum München, Munich, Germany
- Technical University Munich, School of Medicine, Forschergruppe Diabetes at Klinikum rechts der Isar, Munich, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany.
- German Center for Diabetes Research (DZD), Munich, Germany.
- Forschergruppe Diabetes e.V. at Helmholtz Zentrum München, Munich, Germany.
- Technical University Munich, School of Medicine, Forschergruppe Diabetes at Klinikum rechts der Isar, Munich, Germany.
| | - Ezio Bonifacio
- German Center for Diabetes Research (DZD), Munich, Germany
- Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of TU Dresden, Faculty of Medicine, Dresden, Germany
| | | |
Collapse
|
22
|
Hofelich A, Marcus BA, Achenbach P. Früherkennung und Prävention des Typ-1-Diabetes. DIABETOL STOFFWECHS 2022. [DOI: 10.1055/a-0894-1860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
23
|
Stem-Cell-Derived β-Like Cells with a Functional PTPN2 Knockout Display Increased Immunogenicity. Cells 2022; 11:cells11233845. [PMID: 36497105 PMCID: PMC9737324 DOI: 10.3390/cells11233845] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/17/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
Type 1 diabetes is a polygenic disease that results in an autoimmune response directed against insulin-producing beta cells. PTPN2 is a known high-risk type 1 diabetes associated gene expressed in both immune- and pancreatic beta cells, but how genes affect the development of autoimmune diabetes is largely unknown. We employed CRISPR/Cas9 technology to generate a functional knockout of PTPN2 in human pluripotent stem cells (hPSC) followed by differentiating stem-cell-derived beta-like cells (sBC) and detailed phenotypical analyses. The differentiation efficiency of PTPN2 knockout (PTPN2 KO) sBC is comparable to wild-type (WT) control sBC. Global transcriptomics and protein assays revealed the increased expression of HLA Class I molecules in PTPN2 KO sBC at a steady state and upon exposure to proinflammatory culture conditions, indicating a potential for the increased immune recognition of human beta cells upon differential PTPN2 expression. sBC co-culture with autoreactive preproinsulin-reactive T cell transductants confirmed increased immune stimulations by PTPN2 KO sBC compared to WT sBC. Taken together, our results suggest that the dysregulation of PTPN2 expression in human beta cell may prime autoimmune T cell reactivity and thereby contribute to the development of type 1 diabetes.
Collapse
|
24
|
Vehik K, Boulware D, Killian M, Rewers M, McIndoe R, Toppari J, Lernmark Å, Akolkar B, Ziegler AG, Rodriguez H, Schatz DA, Krischer JP, Hagopian W. Rising Hemoglobin A1c in the Nondiabetic Range Predicts Progression of Type 1 Diabetes As Well As Oral Glucose Tolerance Tests. Diabetes Care 2022; 45:2342-2349. [PMID: 36150054 PMCID: PMC9587339 DOI: 10.2337/dc22-0828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/15/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Biomarkers predicting risk of type 1 diabetes (stage 3) among children with islet autoantibodies are greatly needed to prevent diabetic ketoacidosis and facilitate prevention therapies. RESEARCH DESIGN AND METHODS Children in the prospective The Environmental Determinants of Diabetes in the Young (TEDDY) study (n = 707) with confirmed diabetes-associated autoantibodies (GAD antibody, IA-2A, and/or insulin autoantibody) and two or more HbA1c measurements were followed to diabetes or median age 11.1 years. Once confirmed autoantibody positive, HbA1c was measured quarterly. Cox models and receiver operative characteristic curve analyses revealed the prognostic utility for risk of stage 3 on a relative HbA1c increase from the baseline visit or an oral glucose tolerance test (OGTT) 2-h plasma glucose (2-hPG). This HbA1c approach was then validated in the Type 1 Diabetes TrialNet Pathway to Prevention Study (TrialNet) (n = 1,190). RESULTS A 10% relative HbA1c increase from baseline best marked the increased risk of stage 3 in TEDDY (74% sensitive; 88% specific). Significant predictors of risk for HbA1c change were age and HbA1c at the baseline test, genetic sex, maximum number of autoantibodies, and maximum rate of HbA1c increase by time of change. The multivariable model featuring a HbA1c ≥10% increase and these additional factors revealed increased risk of stage 3 in TEDDY (hazard ratio [HR] 12.74, 95% CI 8.7-18.6, P < 0.0001) and TrialNet (HR 5.09, 95% CI 3.3-7.9, P < 0.0001). Furthermore, the composite model using HbA1c ≥10% increase performed similarly to an OGTT 2-hPG composite model (TEDDY area under the curve [AUC] 0.88 and 0.85, respectively) and to the HbA1c model in TrialNet (AUC 0.82). CONCLUSIONS An increase of ≥10% in HbA1c from baseline is as informative as OGTT 2-hPG in predicting risk of stage 3 in youth with genetic risk and diabetes-associated autoantibodies.
Collapse
Affiliation(s)
- Kendra Vehik
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - David Boulware
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL
| | | | - Marian Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO
| | - Richard McIndoe
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA
| | - Jorma Toppari
- Research Centre for Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, and Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University/Clinical Research Centre, Skane University Hospital, Malmö, Sweden
| | - Beena Akolkar
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Anette-G. Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, and Klinikum rechts der Isar, Technische Universität München, and Forschergruppe Diabetes e.V. Neuherberg, Germany
| | - Henry Rodriguez
- USF Diabetes and Endocrinology Center, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Desmond A. Schatz
- Diabetes Center of Excellence, University of Florida, Gainesville, FL
| | - Jeffrey P. Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL
| | | |
Collapse
|
25
|
Abstract
Diabetes is a chronic metabolic disease affecting an increasing number of people. Although diabetes has negative health outcomes for diagnosed individuals, a population at particular risk are pregnant women, as diabetes impacts not only a pregnant woman's health but that of her child. In this review, we cover the current knowledge and unanswered questions on diabetes affecting an expectant mother, focusing on maternal and fetal outcomes.
Collapse
Affiliation(s)
- Cecilia González Corona
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, One Baylor Plaza, Houston, TX 77030, USA
| | - Ronald J. Parchem
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, One Baylor Plaza, Houston, TX 77030, USA,Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
26
|
Badpa M, Wolf K, Schneider A, Winkler C, Haupt F, Peters A, Ziegler AG. Association of long-term environmental exposures in pregnancy and early life with islet autoimmunity development in children in Bavaria, Germany. ENVIRONMENTAL RESEARCH 2022; 212:113503. [PMID: 35609657 DOI: 10.1016/j.envres.2022.113503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/08/2022] [Accepted: 05/16/2022] [Indexed: 05/24/2023]
Abstract
OBJECTIVE Incidence of early-onset type 1 diabetes (T1D) has been increasing worldwide. Only few studies examined the relationship between geographical environmental variation and T1D incidence or its presymptomatic stage of islet autoimmunity. Our study aimed to investigate the effect of long-term environmental exposures during pregnancy and early life on childhood islet autoimmunity. RESEARCH DESIGN AND METHODS We used data from the Fr1da cohort study which screened children aged 1.75-5.99 years for multiple islet autoantibodies in Bavaria, Germany between 2015 and 2019. We included 85,251 children with valid residential information. Daily averages for particulate matter with a diameter <2.5 μm, nitrogen dioxide, ozone, air temperature, and greenness were averaged for each zip-code or directly assigned to the addresses. The exposure windows included pregnancy, the first year and the first two years of life. Generalized additive models adjusting for individual and socioeconomic variables were used to investigate associations between environmental exposures and islet autoimmunity development. RESULTS Islet autoimmunity was diagnosed in 272 children. Colder air temperature during pregnancy was associated with developing islet autoimmunity at the address (per 2.2 °C decrease, Odds ratio (OR): 1.49; 95% Confidence interval (CI): 1.21-1.83) and zip-code level (per 2.4 °C decrease, OR: 1.31; 95% CI: 1.08-1.59). Using the addresses, significant associations were also observed during the first years of life. CONCLUSION In this study, children's residential exposure to lower levels of air temperature during pregnancy and early life increased the risk of islet autoimmunity before the age of six.
Collapse
Affiliation(s)
- Mahnaz Badpa
- Institute of Epidemiology, Helmholtz Zentrum München, Munich, Germany; Institute for Medical Informatics, Biometrics and Epidemiology, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany; German Center for Diabetes Research (DZD), Munich, Germany.
| | - Kathrin Wolf
- Institute of Epidemiology, Helmholtz Zentrum München, Munich, Germany
| | | | - Christiane Winkler
- German Center for Diabetes Research (DZD), Munich, Germany; Institute for Diabetes Research, Helmholtz Diabetes Center at Helmholtz Zentrum München, Munich, Germany
| | - Florian Haupt
- German Center for Diabetes Research (DZD), Munich, Germany; Institute for Diabetes Research, Helmholtz Diabetes Center at Helmholtz Zentrum München, Munich, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, Munich, Germany; German Center for Diabetes Research (DZD), Munich, Germany; Chair of Epidemiology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Anette-Gabriele Ziegler
- German Center for Diabetes Research (DZD), Munich, Germany; Institute for Diabetes Research, Helmholtz Diabetes Center at Helmholtz Zentrum München, Munich, Germany; Forschergruppe Diabetes, Chair of Diabetes and Gestational Diabetes, Faculty of Medicine, Technical University Munich at Klinikum Rechts der Isar, Munich, Germany.
| |
Collapse
|
27
|
Ochsner SA, Pillich RT, Rawool D, Grethe JS, McKenna NJ. Transcriptional regulatory networks of circulating immune cells in type 1 diabetes: A community knowledgebase. iScience 2022; 25:104581. [PMID: 35832893 PMCID: PMC9272393 DOI: 10.1016/j.isci.2022.104581] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/02/2022] Open
Abstract
Investigator-generated transcriptomic datasets interrogating circulating immune cell (CIC) gene expression in clinical type 1 diabetes (T1D) have underappreciated re-use value. Here, we repurposed these datasets to create an open science environment for the generation of hypotheses around CIC signaling pathways whose gain or loss of function contributes to T1D pathogenesis. We firstly computed sets of genes that were preferentially induced or repressed in T1D CICs and validated these against community benchmarks. We then inferred and validated signaling node networks regulating expression of these gene sets, as well as differentially expressed genes in the original underlying T1D case:control datasets. In a set of three use cases, we demonstrated how informed integration of these networks with complementary digital resources supports substantive, actionable hypotheses around signaling pathway dysfunction in T1D CICs. Finally, we developed a federated, cloud-based web resource that exposes the entire data matrix for unrestricted access and re-use by the research community. Re-use of transcriptomic type 1 diabetes (T1D) circulating immune cells (CICs) datasets We generated transcriptional regulatory networks for T1D CICs Use cases generate substantive hypotheses around signaling pathway dysfunction in T1D CICs Networks are freely accessible on the web for re-use by the research community
Collapse
Affiliation(s)
- Scott A. Ochsner
- Department of Molecular, Baylor College of Medicine, Houston, TX 77030, USA
- Cellular Biology and Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rudolf T. Pillich
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Deepali Rawool
- Center for Research in Biological Systems, University of California San Diego, La Jolla, CA 92093, USA
| | - Jeffrey S. Grethe
- Center for Research in Biological Systems, University of California San Diego, La Jolla, CA 92093, USA
| | - Neil J. McKenna
- Department of Molecular, Baylor College of Medicine, Houston, TX 77030, USA
- Cellular Biology and Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Corresponding author
| |
Collapse
|
28
|
Pham VT, Ciccaglione M, Ramirez DG, Benninger RKP. Ultrasound Imaging of Pancreatic Perfusion Dynamics Predicts Therapeutic Prevention of Diabetes in Preclinical Models of Type 1 Diabetes. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1336-1347. [PMID: 35473669 PMCID: PMC9149043 DOI: 10.1016/j.ultrasmedbio.2022.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 02/07/2022] [Accepted: 03/18/2022] [Indexed: 06/14/2023]
Abstract
In type 1 diabetes (T1D), immune-cell infiltration into islets of Langerhans (insulitis) and β-cell decline occur years before diabetes presents. There is a lack of validated clinical approaches for detecting insulitis and β-cell decline, to diagnose eventual diabetes and monitor the efficacy of therapeutic interventions. We previously determined that contrast-enhanced ultrasound measurements of pancreas perfusion dynamics predict disease progression in T1D pre-clinical models. Here, we test whether these measurements predict therapeutic prevention of T1D. We performed destruction-reperfusion measurements with size-isolated microbubbles in non-obese diabetic (NOD)-severe combined immunodeficiency (SCID) mice receiving an adoptive transfer of diabetogenic splenocytes. Mice received vehicle control or the following treatments: (i) anti-CD3 to block T-cell activation; (ii) anti-CD4 to deplete CD4+ T cells; (iii) verapamil to reduce β-cell apoptosis; or (iv) tauroursodeoxycholic acid (TUDCA) to reduce β-cell endoplasmic reticulum stress. We compared measurements of pancreas perfusion dynamics with subsequent progression to diabetes. Anti-CD3, anti-CD4, and verapamil delayed diabetes development. Blood flow dynamics was significantly altered in treated mice with delayed/absent diabetes development compared with untreated mice. Conversely, blood flow dynamics in treated mice with unchanged diabetes development was similar to that in untreated mice. Thus, measurement of pancreas perfusion dynamics predicts the successful prevention of diabetes. This strategy may provide a clinically deployable predictive marker for therapeutic prevention in asymptomatic T1D.
Collapse
Affiliation(s)
- Vinh T Pham
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Mark Ciccaglione
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David G Ramirez
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard K P Benninger
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
29
|
Triolo TM, Pyle L, Broncucia H, Armstrong T, Yu L, Gottlieb PA, Steck AK. Association of High-Affinity Autoantibodies With Type 1 Diabetes High-Risk HLA Haplotypes. J Clin Endocrinol Metab 2022; 107:e1510-e1517. [PMID: 34850014 PMCID: PMC8947772 DOI: 10.1210/clinem/dgab853] [Citation(s) in RCA: 3] [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] [Received: 08/02/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Electrochemiluminescence (ECL) assays are high-affinity autoantibody (Ab) tests that are more specific than Abs detected by traditional radiobinding assays (RBA) for risk screening and prediction of progression to type 1 diabetes. We sought to characterize the association of high-risk human leukocyte antigen (HLA) haplotypes and genotypes with ECL positivity and levels in relatives of individuals with type 1 diabetes. METHODS We analyzed 602 participants from the TrialNet Pathway to Prevention Study who were positive for at least 1 RBA diabetes-related Ab [glutamic acid decarboxylase autoantibodies (GADA) or insulin autoantibodies (IAA)] and for whom ECL and HLA data were available. ECL and RBA Ab levels were converted to SD units away from mean (z-scores) for analyses. RESULTS Mean age at initial visit was 19.4 ± 13.7 years; 344 (57.1%) were female and 104 (17.3%) carried the high-risk HLA-DR3/4*0302 genotype. At initial visit 424/602 (70.4%) participants were positive for either ECL-GADA or ECL-IAA, and 178/602 (29.6%) were ECL negative. ECL and RBA-GADA positivity were associated with both HLA-DR3 and DR4 haplotypes (all Ps < 0.05), while ECL and RBA-GADA z-score titers were higher in participants with HLA-DR3 haplotypes only (both Ps < 0.001). ECL-IAA (but not RBA-IAA) positivity was associated with the HLA-DR4 haplotype (P < 0.05). CONCLUSIONS ECL-GADA positivity is associated with the HLA-DR3 and HLA-DR4 haplotypes and levels are associated with the HLA-DR3 haplotype. ECL-IAA positivity is associated with HLA-DR4 haplotype. These studies further contribute to the understanding of genetic risk and islet autoimmunity endotypes in type 1 diabetes.
Collapse
Affiliation(s)
- Taylor M Triolo
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
- Correspondence: Taylor M. Triolo, MD, University of Colorado Denver School of Medicine, Barbara Davis Center for Diabetes, 1775 Aurora Ct, MS #A140, Aurora, CO, USA 80045-2581.
| | - Laura Pyle
- Department of Pediatrics, University of Colorado, Aurora, CO, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Hali Broncucia
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Taylor Armstrong
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Liping Yu
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Peter A Gottlieb
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Andrea K Steck
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| |
Collapse
|
30
|
Pouresmaeil V, Mashayekhi S, Sarafraz Yazdi M. Investigation of serum level relationship anti-glutamic acid decarboxylase antibody and inflammatory cytokines (IL1-β, IL-6) with vitamins D in type 2 diabetes. J Diabetes Metab Disord 2022; 21:181-187. [PMID: 35673456 PMCID: PMC9167395 DOI: 10.1007/s40200-021-00956-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 12/07/2021] [Indexed: 01/16/2023]
Abstract
Background Various factors are involved in the development and progression of diabetes mellitus, from cytokines and autoimmune destruction of pancreatic beta cells to vitamin D.This study aimed to investigate the association of serum levels of anti-glutamic acid decarboxylase (anti-GAD) and inflammatory cytokines with vitamin D in type 2 diabetes (T2D). Methods This case-control study was performed on 30 patients with T2D and 30 healthy individuals in Mashhad hospitals in 2020. Lipid profile, creatinine, uric acid, FBS, HbA1c, and blood pressure were recorded. All study variables were measured, particularly serum vitamin D, anti-GAD, and inflammatory cytokine levels in diabetic patients, and the data were compared to those from healthy subjects by performing an appropriate statistical analysis. Results Diabetic patients with a mean age of 52.9 ± 10.4 years, including 16 women and healthy individuals with a mean age of 48.5 ± 10.4 years, including 16 women, were studied. BMI level (P = 0.002), systolic blood pressure (P = 0.034), HbA1c, insulin, IL-6, IL1-β, anti-GAD levels, and insulin resistance in diabetic patients were significantly higher than the control group (P = 0.001). The vitamin D level in the control group was significantly higher than in the case group (P = 0.0001). The results showed a significant direct relationship between IL-6, IL-1β, and anti-GAD with HbA1c, FBS, insulin, and insulin resistance. However, there was a significant inverse relationship between IL-6, IL-1β, and anti-GAD with vitamin D. Conclusions Inflammatory cytokines and anti-GAD and vitamin D are associated with diabetes, and thus controlling these factors can help improve T2D.
Collapse
Affiliation(s)
- Vahid Pouresmaeil
- grid.411768.d0000 0004 1756 1744Department of Biochemistry, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran ,grid.411768.d0000 0004 1756 1744Innovative Medical Research Center, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran ,grid.411768.d0000 0004 1756 1744Faculty of Medicine Shahinfar, Islamic Azad University, Sarab Street, Mashhad, Iran
| | - Sarmad Mashayekhi
- grid.411768.d0000 0004 1756 1744Department of Biology, Islamic Azad University, Mashhad Branch, Mashhad, Iran
| | - Mohammad Sarafraz Yazdi
- grid.411768.d0000 0004 1756 1744Department of Internal Medicine, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| |
Collapse
|
31
|
Ravi K, Pasi R. Type 1 diabetes mellitus in pediatric age group: A rising endemic. J Family Med Prim Care 2022; 11:27-31. [PMID: 35309606 PMCID: PMC8930152 DOI: 10.4103/jfmpc.jfmpc_975_21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 11/04/2022] Open
Abstract
Type 1 Diabetes Mellitus is the most common endocrinological abnormality found in children. The incidence of T1DM has steadily increased in nearly all parts of the world. Both genetic susceptibility and environmental factors contribute to the pathogenesis. It is caused due to either decreased or absent insulin production in the body due to multiple etiologies. We have done a literature review of type 1 DM in children and a clinical audit of point prevalence of type 1 DM cases & its clinical correlates of patients presenting at the tertiary level hospital, AIIMS Rishikesh, over two years six months period (April 2015 to September 2017). We found the prevalence of diabetes mellitus (Type 1) is 2.88%. Among clinical features at presentation, 56.5% presented with polyuria, 34.8% with polydipsia, 21.7% with polyphagia, 39.1% with weight loss. 26.1% of patients had diabetic ketoacidosis at presentation. The majority of children have deranged HbA1C levels (94.4%). It is concluded that children presented with higher HbA1c levels at onset and higher duration of symptoms are at greater risk for the development of complications.
Collapse
|
32
|
Yazdanpanah N, Yazdanpanah M, Wang Y, Forgetta V, Pollak M, Polychronakos C, Richards JB, Manousaki D. Clinically Relevant Circulating Protein Biomarkers for Type 1 Diabetes: Evidence From a Two-Sample Mendelian Randomization Study. Diabetes Care 2022; 45:169-177. [PMID: 34758976 DOI: 10.2337/dc21-1049] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 10/18/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To identify circulating proteins influencing type 1 diabetes susceptibility using Mendelian randomization (MR). RESEARCH DESIGN AND METHODS We used a large-scale two-sample MR study, using cis genetic determinants (protein quantitative trait loci [pQTL]) of up to 1,611 circulating proteins from five large genome-wide association studies, to screen for causal associations of these proteins with type 1 diabetes risk in 9,684 case subjects with type 1 diabetes and 15,743 control subjects. Further, pleiotropy-robust MR methods were used in sensitivity analyses using both cis and trans-pQTL. RESULTS We found that a genetically predicted SD increase in signal regulatory protein gamma (SIRPG) level was associated with increased risk of type 1 diabetes risk (MR odds ratio [OR] 1.66 [95% 1.36-2.03]; P = 7.1 × 10-7). The risk of type 1 diabetes increased almost twofold per genetically predicted standard deviation (SD) increase in interleukin-27 Epstein-Barr virus-induced 3 (IL27-EBI3) protein levels (MR OR 1.97 [95% CI 1.48-2.62]; P = 3.7 × 10-6). However, an SD increase in chymotrypsinogen B1 (CTRB1) was associated with decreased risk of type 1 diabetes (MR OR 0.84 [95% CI 0.77-0.90]; P = 6.1 × 10-6). Sensitivity analyses using MR methods testing for pleiotropy while including trans-pQTL showed similar results. While the MR-Egger suggested no pleotropic effect (P value MR-Egger intercept = 0.31), there was evidence of pleiotropy in MR-PRESSO (P value global test = 0.006). CONCLUSIONS We identified three novel circulating protein biomarkers associated with type 1 diabetes risk using an MR approach. These biomarkers are promising targets for development of drugs and/or of screening tools for early prediction of type 1 diabetes.
Collapse
Affiliation(s)
- Nahid Yazdanpanah
- 1Research Center of the Sainte-Justine University Hospital, University of Montreal, Montreal, Quebec, Canada
| | - Mojgan Yazdanpanah
- 1Research Center of the Sainte-Justine University Hospital, University of Montreal, Montreal, Quebec, Canada
| | - Ye Wang
- 2Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Vincenzo Forgetta
- 2Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Michael Pollak
- 2Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.,3Department of Medicine, McGill University, Montreal, Quebec, Canada.,4Department of Oncology, McGill University, Montreal, Quebec, Canada
| | - Constantin Polychronakos
- 5Department of Pediatrics, McGill University, Montreal, Quebec, Canada.,6Department of Human Genetics, McGill University, Montreal, Quebec, Canada.,7Centre of Excellence in Translational Immunology, Montreal, Quebec, Canada
| | - J Brent Richards
- 2Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.,3Department of Medicine, McGill University, Montreal, Quebec, Canada.,6Department of Human Genetics, McGill University, Montreal, Quebec, Canada.,8Department of Epidemiology and Biostatistics, McGill University, Montreal, Quebec, Canada.,9Department of Twin Research, King's College London, London, U.K
| | - Despoina Manousaki
- 1Research Center of the Sainte-Justine University Hospital, University of Montreal, Montreal, Quebec, Canada.,10Departments of Pediatrics, Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
33
|
Silverstein A, Dudaev A, Studneva M, Aitken J, Blokh S, Miller AD, Tanasova S, Rose N, Ryals J, Borchers C, Nordstrom A, Moiseyakh M, Herrera AS, Skomorohov N, Marshall T, Wu A, Cheng RH, Syzko K, Cotter PD, Podzyuban M, Thilly W, Smith PD, Barach P, Bouri K, Schoenfeld Y, Matsuura E, Medvedeva V, Shmulevich I, Cheng L, Seegers P, Khotskaya Y, Flaherty K, Dooley S, Sorenson EJ, Ross M, Suchkov S. Evolution of biomarker research in autoimmunity conditions for health professionals and clinical practice. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 190:219-276. [DOI: 10.1016/bs.pmbts.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
34
|
Abstract
INTRODUCTION Despite advances in technology including the development of more sophisticated methods of monitoring blood glucose and delivering insulin, many individuals with type 1 diabetes continue to experience significant challenges in optimizing glycaemic control. Alternative treatment approaches to insulin are required. Increasing efforts have focused on developing treatments aimed at targeting the underlying disease process to modulate the immune system, maximize beta cell function and enhance endogenous insulin production and action. SOURCES OF DATA Literature searches with keywords 'Type 1 diabetes and immunotherapy', publications relating to clinical trials of immunotherapy in type 1 diabetes. AREAS OF AGREEMENT Insulin therapy is insufficient to achieve optimal glycaemic control in many individuals with type 1 diabetes, and new treatment approaches are required. Studies have showed promising results for the use of immunotherapy as a means of delaying disease onset and progression. AREAS OF CONTROVERSY The optimal way of identifying individuals most likely to benefit from immunotherapies. GROWING POINTS A better understanding of the natural history of type 1 diabetes has made it possible to identify individuals who have developed autoimmunity but have not yet progressed to clinical diabetes, offering opportunities not only to develop treatments that delay disease progression, but prevent its development in the first place. A consensus on how to identify individuals who may benefit from immunotherapy to prevent disease onset is needed. AREAS TIMELY FOR DEVELOPING RESEARCH The development of optimal strategies for preventing and delaying progression of type 1 diabetes, and monitoring the response to immunointervention.
Collapse
Affiliation(s)
- L A Allen
- Diabetes Research Group, Cardiff University, Cardiff, UK
| | - C M Dayan
- Diabetes Research Group, Cardiff University, Cardiff, UK
| |
Collapse
|
35
|
Vallianou NG, Stratigou T, Geladari E, Tessier CM, Mantzoros CS, Dalamaga M. Diabetes type 1: Can it be treated as an autoimmune disorder? Rev Endocr Metab Disord 2021; 22:859-876. [PMID: 33730229 DOI: 10.1007/s11154-021-09642-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/03/2021] [Indexed: 02/06/2023]
Abstract
Type 1 Diabetes Mellitus (T1DM) is characterized by progressive autoimmune-mediated destruction of the pancreatic beta-cells leading to insulin deficiency and hyperglycemia. It is associated with significant treatment burden and necessitates life-long insulin therapy. The role of immunotherapy in the prevention and management of T1DM is an evolving area of interest which has the potential to alter the natural history of this disease.In this review, we give insight into recent clinical trials related to the use of immunotherapeutic approaches for T1DM, such as proinflammatory cytokine inhibition, cell-depletion and cell-therapy approaches, autoantigen-specific treatments and stem cell therapies. We highlight the timing of intervention, aspects of therapy including adverse effects and the emergence of a novel lymphocyte crucial in T1DM autoimmunity. We also discuss the role of cardiac autoimmunity and its link to excess CVD risk in T1DM.We conclude that significant advances have been made in development of immunotherapeutic targets and agents for the treatment and prevention of T1DM. These immune-based therapies promise preservation of beta-cells and decreasing insulin dependency. In their current state, immunotherapeutic approaches cannot yet halt the progression from a preclinical state to overt T1DM nor can they replace standard insulin therapy in existing T1DM. It remains to be seen whether immunotherapy will ultimately play a key role in the prevention of progression to overt T1DM and whether it may find a place in our therapeutic armamentarium to improve clinical outcomes and quality of life in established T1DM.
Collapse
Affiliation(s)
- Natalia G Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Theodora Stratigou
- Department of Endocrinology, Diabetes and Metabolic Diseases, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527, Athens, Goudi, Greece
| | - Eleni Geladari
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou str, 10676, Athens, Greece
| | - Christopher M Tessier
- Endocrinology Section, VA Boston Healthcare System, 1400 VFW Parkway West Roxbury, Boston, MA, 02132, USA.
| | - Christos S Mantzoros
- Endocrinology Section, VA Boston Healthcare System, 1400 VFW Parkway West Roxbury, Boston, MA, 02132, USA
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527, Athens, Goudi, Greece
| |
Collapse
|
36
|
Korneva KG, Strongin LG, Kolbasina EV, Budylina MV, Makeeva NV, Zagainov VE. Diagnostic Capabilities of Islet Autoantibodies in Children with New-Onset Type 1 Diabetes Mellitus and Healthy Siblings. Sovrem Tekhnologii Med 2021; 12:29-34. [PMID: 34796016 PMCID: PMC8596233 DOI: 10.17691/stm2020.12.6.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Indexed: 12/16/2022] Open
Abstract
The aim of the study is to determine the diagnostic utility of several islet autoantibodies and their combinations in order to identify individuals susceptible to type 1 diabetes mellitus (T1DM) among healthy siblings in the pediatric population within the scope of the development of a screening program.
Collapse
Affiliation(s)
- K G Korneva
- Associate Professor, Department of Endocrinology and Internal Medicine; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - L G Strongin
- Professor, Head of the Department of Endocrinology and Internal Medicine; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - E V Kolbasina
- Pediatric Endocrinologist, Head of the Department of Endocrinology; Nizhny Novgorod Regional Children's Clinical Hospital, 211 Vaneeva St., Nizhny Novgorod, 603136, Russia
| | - M V Budylina
- Head of the Department of Pediatric Endocrinology and Gastroenterology; Republican Children's Clinical Hospital of the Ministry of Health of the Chuvash Republic, 27 Fedora Gladkova St., Cheboksary, 428020, Russia
| | - N V Makeeva
- Pediatric Endocrinologist, Chief Non-Staff Pediatric Endocrinologist; Children's Republican Clinical Hospital, 10 Meditsinskaya St., Yoshkar-Ola, the Republic of Mari El, 424005, Russia
| | - V E Zagainov
- Associate Professor, Head of the Department of Faculty Surgery and Transplantology Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| |
Collapse
|
37
|
Detecting insulitis in type 1 diabetes with ultrasound phase-change contrast agents. Proc Natl Acad Sci U S A 2021; 118:2022523118. [PMID: 34607942 DOI: 10.1073/pnas.2022523118] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2021] [Indexed: 11/18/2022] Open
Abstract
Type 1 diabetes (T1D) results from immune infiltration and destruction of insulin-producing β cells within the pancreatic islets of Langerhans (insulitis). Early diagnosis during presymptomatic T1D would allow for therapeutic intervention prior to substantial β-cell loss at onset. There are limited methods to track the progression of insulitis and β-cell mass decline. During insulitis, the islet microvasculature increases permeability, such that submicron-sized particles can extravasate and accumulate within the islet microenvironment. Ultrasound is a widely deployable and cost-effective clinical imaging modality. However, conventional microbubble contrast agents are restricted to the vasculature. Submicron nanodroplet (ND) phase-change agents can be vaporized into micron-sized bubbles, serving as a microbubble precursor. We tested whether NDs extravasate into the immune-infiltrated islet microenvironment. We performed ultrasound contrast-imaging following ND infusion in nonobese diabetic (NOD) mice and NOD;Rag1ko controls and tracked diabetes development. We measured the biodistribution of fluorescently labeled NDs, with histological analysis of insulitis. Ultrasound contrast signal was elevated in the pancreas of 10-wk-old NOD mice following ND infusion and vaporization but was absent in both the noninfiltrated kidney of NOD mice and the pancreas of Rag1ko controls. High-contrast elevation also correlated with rapid diabetes onset. Elevated contrast was also observed as early as 4 wk, prior to mouse insulin autoantibody detection. In the pancreata of NOD mice, infiltrated islets and nearby exocrine tissue were selectively labeled with fluorescent NDs. Thus, contrast ultrasound imaging with ND phase-change agents can detect insulitis prior to diabetes onset. This will be important for monitoring disease progression, to guide and assess preventative therapeutic interventions for T1D.
Collapse
|
38
|
Nguyen P, Ohnmacht AJ, Galhoz A, Büttner M, Theis F, Menden MP. Künstliche Intelligenz und maschinelles Lernen in der Diabetesforschung. DIABETOLOGE 2021. [DOI: 10.1007/s11428-021-00817-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
39
|
Kahn SE, Chen YC, Esser N, Taylor AJ, van Raalte DH, Zraika S, Verchere CB. The β Cell in Diabetes: Integrating Biomarkers With Functional Measures. Endocr Rev 2021; 42:528-583. [PMID: 34180979 PMCID: PMC9115372 DOI: 10.1210/endrev/bnab021] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Indexed: 02/08/2023]
Abstract
The pathogenesis of hyperglycemia observed in most forms of diabetes is intimately tied to the islet β cell. Impairments in propeptide processing and secretory function, along with the loss of these vital cells, is demonstrable not only in those in whom the diagnosis is established but typically also in individuals who are at increased risk of developing the disease. Biomarkers are used to inform on the state of a biological process, pathological condition, or response to an intervention and are increasingly being used for predicting, diagnosing, and prognosticating disease. They are also proving to be of use in the different forms of diabetes in both research and clinical settings. This review focuses on the β cell, addressing the potential utility of genetic markers, circulating molecules, immune cell phenotyping, and imaging approaches as biomarkers of cellular function and loss of this critical cell. Further, we consider how these biomarkers complement the more long-established, dynamic, and often complex measurements of β-cell secretory function that themselves could be considered biomarkers.
Collapse
Affiliation(s)
- Steven E Kahn
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - Yi-Chun Chen
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Nathalie Esser
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - Austin J Taylor
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Daniël H van Raalte
- Department of Internal Medicine, Amsterdam University Medical Center (UMC), Vrije Universiteit (VU) University Medical Center, 1007 MB Amsterdam, The Netherlands.,Department of Experimental Vascular Medicine, Amsterdam University Medical Center (UMC), Academic Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Sakeneh Zraika
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, VA Puget Sound Health Care System and University of Washington, Seattle, 98108 WA, USA
| | - C Bruce Verchere
- BC Children's Hospital Research Institute and Centre for Molecular Medicine and Therapeutics, Vancouver, BC, V5Z 4H4, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| |
Collapse
|
40
|
Krupa A, Kowalska I. The Kynurenine Pathway-New Linkage between Innate and Adaptive Immunity in Autoimmune Endocrinopathies. Int J Mol Sci 2021; 22:9879. [PMID: 34576041 PMCID: PMC8469440 DOI: 10.3390/ijms22189879] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/18/2022] Open
Abstract
The kynurenine pathway (KP) is highly regulated in the immune system, where it promotes immunosuppression in response to infection or inflammation. Indoleamine 2,3-dioxygenase 1 (IDO1), the main enzyme of KP, has a broad spectrum of activity on immune cells regulation, controlling the balance between stimulation and suppression of the immune system at sites of local inflammation, relevant to a wide range of autoimmune and inflammatory diseases. Various autoimmune diseases, among them endocrinopathies, have been identified to date, but despite significant progress in their diagnosis and treatment, they are still associated with significant complications, morbidity, and mortality. The precise cellular and molecular mechanisms leading to the onset and development of autoimmune disease remain poorly clarified so far. In breaking of tolerance, the cells of the innate immunity provide a decisive microenvironment that regulates immune cells' differentiation, leading to activation of adaptive immunity. The current review provided a comprehensive presentation of the known role of IDO1 and KP activation in the regulation of the innate and adaptive arms of the immune system. Significant attention has been paid to the immunoregulatory role of IDO1 in the most prevalent, organ-specific autoimmune endocrinopathies-type 1 diabetes mellitus (T1DM) and autoimmune thyroiditis.
Collapse
Affiliation(s)
- Anna Krupa
- Department of Internal Medicine and Metabolic Diseases, Medical University of Bialystok, M. Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
| | - Irina Kowalska
- Department of Internal Medicine and Metabolic Diseases, Medical University of Bialystok, M. Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland
| |
Collapse
|
41
|
Anand V, Li Y, Liu B, Ghalwash M, Koski E, Ng K, Dunne JL, Jönsson J, Winkler C, Knip M, Toppari J, Ilonen J, Killian MB, Frohnert BI, Lundgren M, Ziegler AG, Hagopian W, Veijola R, Rewers M. Islet Autoimmunity and HLA Markers of Presymptomatic and Clinical Type 1 Diabetes: Joint Analyses of Prospective Cohort Studies in Finland, Germany, Sweden, and the U.S. Diabetes Care 2021; 44:dc201836. [PMID: 34162665 PMCID: PMC8929180 DOI: 10.2337/dc20-1836] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 04/07/2021] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To combine prospective cohort studies, by including HLA harmonization, and estimate risk of islet autoimmunity and progression to clinical diabetes. RESEARCH DESIGN AND METHODS For prospective cohorts in Finland, Germany, Sweden, and the U.S., 24,662 children at increased genetic risk for development of islet autoantibodies and type 1 diabetes have been followed. Following harmonization, the outcomes were analyzed in 16,709 infants-toddlers enrolled by age 2.5 years. RESULTS In the infant-toddler cohort, 1,413 (8.5%) developed at least one autoantibody confirmed at two or more consecutive visits (seroconversion), 865 (5%) developed multiple autoantibodies, and 655 (4%) progressed to diabetes. The 15-year cumulative incidence of diabetes varied in children with one, two, or three autoantibodies at seroconversion: 45% (95% CI 40-52), 85% (78-90), and 92% (85-97), respectively. Among those with a single autoantibody, status 2 years after seroconversion predicted diabetes risk: 12% (10-25) if reverting to autoantibody negative, 30% (20-40) if retaining a single autoantibody, and 82% (80-95) if developing multiple autoantibodies. HLA-DR-DQ affected the risk of confirmed seroconversion and progression to diabetes in children with stable single-autoantibody status. Their 15-year diabetes incidence for higher- versus lower-risk genotypes was 40% (28-50) vs. 12% (5-38). The rate of progression to diabetes was inversely related to age at development of multiple autoantibodies, ranging from 20% per year to 6% per year in children developing multipositivity in ≤2 years or >7.4 years, respectively. CONCLUSIONS The number of islet autoantibodies at seroconversion reliably predicts 15-year type 1 diabetes risk. In children retaining a single autoantibody, HLA-DR-DQ genotypes can further refine risk of progression.
Collapse
Affiliation(s)
- Vibha Anand
- Center for Computational Health, IBM T.J. Watson Research Center, Cambridge, MA, and Yorktown Heights, NY
| | - Ying Li
- Center for Computational Health, IBM T.J. Watson Research Center, Cambridge, MA, and Yorktown Heights, NY
| | - Bin Liu
- Center for Computational Health, IBM T.J. Watson Research Center, Cambridge, MA, and Yorktown Heights, NY
| | - Mohamed Ghalwash
- Center for Computational Health, IBM T.J. Watson Research Center, Cambridge, MA, and Yorktown Heights, NY
- Ain Shams University, Cairo, Egypt
| | - Eileen Koski
- Center for Computational Health, IBM T.J. Watson Research Center, Cambridge, MA, and Yorktown Heights, NY
| | - Kenney Ng
- Center for Computational Health, IBM T.J. Watson Research Center, Cambridge, MA, and Yorktown Heights, NY
| | | | - Josefine Jönsson
- Department of Clinical Sciences Malmö, Lund University/CRC, Skåne University Hospital, Malmö
| | - Christiane Winkler
- Institute of Diabetes Research, Helmholtz Zentrum München German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes e.V. am Helmholtz Zentrum München, Munich, Germany
- Forschergruppe Diabetes, Technical University Munich, Germany
| | - Mikael Knip
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland
- Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Jorma Toppari
- Institute of Biomedicine and Population Research Centre, University of Turku, and Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, and Clinical Microbiology, Turku University Hospital, Turku, Finland
| | | | | | - Markus Lundgren
- Institute of Diabetes Research, Helmholtz Zentrum München German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München German Research Center for Environmental Health, Munich-Neuherberg, Germany
- Forschergruppe Diabetes e.V. am Helmholtz Zentrum München, Munich, Germany
- Forschergruppe Diabetes, Technical University Munich, Germany
| | | | - Riitta Veijola
- PEDEGO Research Unit, Department of Pediatrics, University of Oulu and Oulu University Hospital, Oulu, Finland
| | | |
Collapse
|
42
|
Tangjittipokin W, Umjai P, Khemaprasit K, Charoentawornpanich P, Chanprasert C, Teerawattanapong N, Narkdontri T, Santiprabhob J. Vitamin D pathway gene polymorphisms, vitamin D level, and cytokines in children with type 1 diabetes. Gene 2021; 791:145691. [PMID: 33961971 DOI: 10.1016/j.gene.2021.145691] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/05/2021] [Accepted: 04/30/2021] [Indexed: 02/06/2023]
Abstract
AIMS The study aimed to examine genetic polymorphism of vitamin D-related genes and association between those genes and vitamin D and cytokines levels in children with type 1 diabetes (T1D). MATERIALS AND METHODS This study was conducted among 100 T1D children and 100 controls at Division of Endocrinology and Metabolism, Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand, during 2016 to 2018. Vitamin D metabolite levels were measured by liquid chromatography-tandem mass spectrometry method, serum cytokine levels of IFN- ɣ, IL-10, IL-13, IL-17α, IL-2, IL-4, IL-6, and TNF-α by immunoassay, and genetic variations at VDR, CYP2R1, CYP27B1, GC, DHCR7, and CYP24A1 by polymerase chain reaction-restriction fragment length polymorphism method. RESULTS A relationship between studied single nucleotide polymorphisms and T1D was found in CYP2R1 (rs10741657) (GA, OR: 1.83, 95% CI: 1.01-3.31; p = 0.04). VDR haplotypes were also remarkably different between T1D patients and controls. Controls had higher frequency of haplotype TACT than T1D patients (p = 0.02). Vitamin D and all cytokine levels, except for IL-17α, were significantly increased in T1D compared to controls. The polymorphism of DHCR7 (rs12785878) was positively associated with 25OHD3 and 3epi25OHD3 levels and was negatively associated with 25OHD2 level. On the other hand, polymorphism of CYP27B1 (rs4646536) was negatively associated with 3epi25OHD3 level. Polymorphisms of CYP27B1 (rs4646536) and GC (rs2282679) were positively associated with TNF-α levels. VDR variation of rs1544410, rs731236, and rs7975232 also showed negative association with IL-10 levels. In contrast, the level of IL-10 was positively associated with DHCR7 (rs12785878). CONCLUSION Relationships between T1D and CYP2R1 polymorphism and VDR haplotype were found. Vitamin D gene-related variations were associated with vitamin D and circulating cytokine levels in children with T1D.
Collapse
Affiliation(s)
- Watip Tangjittipokin
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Center of Research Excellence for Diabetes and Obesity, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pichakorn Umjai
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Khwanhatai Khemaprasit
- Siriraj Diabetes Center of Excellence, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Parichat Charoentawornpanich
- Division of Endocrinology and Metabolism, Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chutima Chanprasert
- Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nipaporn Teerawattanapong
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Center of Research Excellence for Diabetes and Obesity, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tassanee Narkdontri
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Siriraj Center of Research Excellence for Diabetes and Obesity, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jeerunda Santiprabhob
- Siriraj Diabetes Center of Excellence, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Endocrinology and Metabolism, Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
43
|
Suneja S, Gangopadhyay S, Saini V, Dawar R, Kaur C. Emerging Diabetic Novel Biomarkers of the 21st Century. ANNALS OF THE NATIONAL ACADEMY OF MEDICAL SCIENCES (INDIA) 2021. [DOI: 10.1055/s-0041-1726613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AbstractDiabetes is a growing epidemic with estimated prevalence of infected to reach ~592 million by the year 2035. An effective way to approach is to detect the disease at a very early stage to reduce the complications and improve lifestyle management. Although several traditional biomarkers including glucated hemoglobin, glucated albumin, fructosamine, and 1,5-anhydroglucitol have helped in ease of diagnosis, there is lack of sensitivity and specificity and are inaccurate in certain clinical settings. Thus, search for new and effective biomarkers is a continuous process with an aim of accurate and timely diagnosis. Several novel biomarkers have surged in the present century that are helpful in timely detection of the disease condition. Although it is accepted that a single biomarker will have its inherent limitations, combining several markers will help to identify individuals at high risk of developing prediabetes and eventually its progression to frank diabetes. This review describes the novel biomarkers of the 21st century, both in type 1 and type 2 diabetes mellitus, and their present potential for assessing risk stratification due to insulin resistance that will pave the way for improved clinical outcome.
Collapse
Affiliation(s)
- Shilpa Suneja
- Department of Biochemistry, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Sukanya Gangopadhyay
- Department of Biochemistry, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Vandana Saini
- Department of Biochemistry, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Rajni Dawar
- Department of Biochemistry, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Charanjeet Kaur
- Department of Biochemistry, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| |
Collapse
|
44
|
Xhonneux LP, Knight O, Lernmark Å, Bonifacio E, Hagopian WA, Rewers MJ, She JX, Toppari J, Parikh H, Smith KGC, Ziegler AG, Akolkar B, Krischer JP, McKinney EF. Transcriptional networks in at-risk individuals identify signatures of type 1 diabetes progression. Sci Transl Med 2021; 13:eabd5666. [PMID: 33790023 PMCID: PMC8447843 DOI: 10.1126/scitranslmed.abd5666] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/24/2020] [Accepted: 03/12/2021] [Indexed: 12/11/2022]
Abstract
Type 1 diabetes (T1D) is a disease of insulin deficiency that results from autoimmune destruction of pancreatic islet β cells. The exact cause of T1D remains unknown, although asymptomatic islet autoimmunity lasting from weeks to years before diagnosis raises the possibility of intervention before the onset of clinical disease. The number, type, and titer of islet autoantibodies are associated with long-term disease risk but do not cause disease, and robust early predictors of individual progression to T1D onset remain elusive. The Environmental Determinants of Diabetes in the Young (TEDDY) consortium is a prospective cohort study aiming to determine genetic and environmental interactions causing T1D. Here, we analyzed longitudinal blood transcriptomes of 2013 samples from 400 individuals in the TEDDY study before both T1D and islet autoimmunity. We identified and interpreted age-associated gene expression changes in healthy infancy and age-independent changes tracking with progression to both T1D and islet autoimmunity, beginning before other evidence of islet autoimmunity was present. We combined multivariate longitudinal data in a Bayesian joint model to predict individual risk of T1D onset and validated the association of a natural killer cell signature with progression and the model's predictive performance on an additional 356 samples from 56 individuals in the independent Type 1 Diabetes Prediction and Prevention study. Together, our results indicate that T1D is characterized by early and longitudinal changes in gene expression, informing the immunopathology of disease progression and facilitating prediction of its course.
Collapse
Affiliation(s)
- Louis-Pascal Xhonneux
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Oliver Knight
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University/CRC Skåne University Hospital Malmo, Jan Waldenströms gata 35, Malmö, Sweden
| | - Ezio Bonifacio
- Center for Regenerative Therapies, Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany
| | - William A Hagopian
- Pacific Northwest Research Institute, 720 Broadway, Seattle, WA 98122, USA
| | - Marian J Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado, 1775 Aurora Ct, Aurora, CO 80045, USA
| | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, 1462 Laney Walker Blvd., Augusta, GA 30912, USA
| | - Jorma Toppari
- Department of Pediatrics, Turku University Hospital, Kiinamyllynkatu 4-8, 20521 Turku, Finland
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, FI-20014 Turun Lyliopisto, Finland
| | - Hemang Parikh
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
| | - Anette-G Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, and Klinikum rechts der Isar, Technische, Universität München, Forschergruppe Diabetes e.V., Arcisstraße 21, 80333 München, Germany
| | - Beena Akolkar
- National Institute of Diabetes and Digestive and Kidney Diseases, 9000 Rockville Pike Bethesda, MD 20892, USA
| | - Jeffrey P Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Eoin F McKinney
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0QQ, UK
- Cambridge Centre for Artificial Intelligence in Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
45
|
Burbelo PD, Iadarola MJ, Keller JM, Warner BM. Autoantibodies Targeting Intracellular and Extracellular Proteins in Autoimmunity. Front Immunol 2021; 12:548469. [PMID: 33763057 PMCID: PMC7982651 DOI: 10.3389/fimmu.2021.548469] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022] Open
Abstract
Detecting autoantibodies provides foundational information for the diagnosis of most autoimmune diseases. An important pathophysiological distinction is whether autoantibodies are directed against extracellular or intracellular proteins. Autoantibodies targeting extracellular domains of proteins, such as membrane receptors, channels or secreted molecules are often directly pathogenic, whereby autoantibody binding to the autoantigen disrupts the normal function of a critical protein or pathway, and/or triggers antibody-dependent cell surface complement killing. By comparison, autoantibodies directed against intracellular proteins are recognized as useful diagnostic biomarkers of abnormal autoimmune activity, but the link between antigenicity and pathogenicity is less straightforward. Because intracellular autoantigens are generally inaccessible to autoantibody binding, for the most part, they do not directly contribute to pathogenesis. In a few diseases, autoantibodies to intracellular targets cause damage indirectly by immune complex formation, immune activation, and other processes. In this review, the general features of and differences between autoimmune diseases segregated on the basis of intracellular or extracellular autoantigens are explored using over twenty examples. Expression profiles of autoantigens in relation to the tissues targeted by autoimmune disease and the temporal appearance of autoantibodies before clinical diagnosis often correlate with whether the respective autoantibodies mostly recognize either intracellular or extracellular autoantigens. In addition, current therapeutic strategies are discussed from this vantage point. One drug, rituximab, depletes CD20+ B-cells and is highly effective for autoimmune disorders associated with autoantibodies against extracellular autoantigens. In contrast, diseases associated with autoantibodies directed predominately against intracellular autoantigens show much more complex immune cell involvement, such as T-cell mediated tissue damage, and require different strategies for optimal therapeutic benefit. Understanding the clinical ramifications of autoimmunity derived by autoantibodies against either intracellular or extracellular autoantigens, or a spectrum of both, has practical implications for guiding drug development, generating monitoring tools, stratification of patient interventions, and designing trials based on predictive autoantibody profiles for autoimmune diseases.
Collapse
Affiliation(s)
- Peter D Burbelo
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Jason M Keller
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Blake M Warner
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
46
|
Triolo TM, Pyle L, Seligova S, Yu L, Simmons K, Gottlieb P, Evans-Molina C, Steck AK. Proinsulin:C-peptide ratio trajectories over time in relatives at increased risk of progression to type 1 diabetes. J Transl Autoimmun 2021; 4:100089. [PMID: 33748733 PMCID: PMC7972972 DOI: 10.1016/j.jtauto.2021.100089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/04/2021] [Accepted: 02/12/2021] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Biomarkers are needed to characterize heterogeneity within populations at risk for type 1 diabetes. The ratio of proinsulin to C-peptide (PI:C ratio), has been proposed as a biomarker of beta cell dysfunction and is associated with progression to type 1 diabetes. However, relationships between PI:C ratios and autoantibody type and number have not been examined. We sought to characterize PI:C ratios in multiple islet autoantibody positive, single autoantibody positive and autoantibody negative relatives of individuals with type 1 diabetes. METHODS We measured PI:C ratios and autoantibodies with both electrochemiluminescence (ECL) assays (ECL-IAA, ECL-GADA and ECL-IA2A) and radiobinding (RBA) assays (mIAA, GADA, IA2A and ZnT8A) in 98 relatives of individuals with type 1 diabetes followed in the TrialNet Pathway to Prevention Study at the Barbara Davis Center for a mean of 7.4 ± 4.1 years. Of these subjects, eight progressed to T1D, 31 were multiple autoantibody (Ab) positive, 37 were single Ab positive and 22 were Ab negative (by RBA). RESULTS In cross-sectional analyses, there were no significant differences in PI:C ratios between type 1 diabetes and/or multiple Ab positive subjects (4.16 ± 4.06) compared to single Ab positive subjects (4.08 ± 4.34) and negative Ab subjects (3.72 ± 3.78) (p = 0.92) overall or after adjusting for age, sex and BMI. Higher PI:C ratios were associated with mIAA titers (p = 0.03) and showed an association with ECL-IA2A titers (p = 0.09), but not with ECL-IAA, GADA, ECL-GADA, IA2A nor ZnT8A titers. In mixed-effects longitudinal models, the trajectories of PI:C ratio over time were significantly different between the Ab negative and multiple Ab positive/type 1 diabetes groups, after adjusting for sex, age, and BMI (p = 0.04). CONCLUSIONS PI:C ratio trajectories increase over time in subjects who have multiple Ab or develop type 1 diabetes and may be a helpful biomarker to further characterize and stratify risk of progression to type 1 diabetes over time.
Collapse
Affiliation(s)
- Taylor M Triolo
- University of Colorado Denver School of Medicine - the Barbara Davis Center for Diabetes, Aurora, CO, USA
| | - Laura Pyle
- University of Colorado Denver School of Medicine - the Barbara Davis Center for Diabetes, Aurora, CO, USA.,University of Colorado Anschutz Medical Campus, Pediatrics, Aurora, CO, USA
| | - Sona Seligova
- University of Colorado Denver School of Medicine - the Barbara Davis Center for Diabetes, Aurora, CO, USA
| | - Liping Yu
- University of Colorado Denver School of Medicine - the Barbara Davis Center for Diabetes, Aurora, CO, USA
| | - Kimber Simmons
- University of Colorado Denver School of Medicine - the Barbara Davis Center for Diabetes, Aurora, CO, USA
| | - Peter Gottlieb
- University of Colorado Denver School of Medicine - the Barbara Davis Center for Diabetes, Aurora, CO, USA
| | - Carmella Evans-Molina
- Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana University Center for Diabetes and Metabolic Diseases. Indianapolis, IN, USA
| | - Andrea K Steck
- University of Colorado Denver School of Medicine - the Barbara Davis Center for Diabetes, Aurora, CO, USA
| |
Collapse
|
47
|
Cepon-Robins TJ. Measuring attack on self: The need for field-friendly methods development and research on autoimmunity in human biology. Am J Hum Biol 2020; 33. [PMID: 33289250 DOI: 10.1002/ajhb.23544] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Autoimmune and inflammatory disorder (AIID) prevalence appears to be increasing in all but the world's poorest regions and countries. Autoimmune diseases occur when there is a breakdown in processes that regulate inflammation and self-recognition by immune cells. Very few field-based studies have been conducted among Indigenous populations and underserved communities with limited access to medical care. This is due, in part, to the fact that autoimmune diseases are difficult to diagnose, even in clinical settings. In remote field settings these difficulties are compounded by the absence of infrastructure necessary for sample storage and analysis, and the lack of hospital/clinic access for more invasive diagnostic procedures. Because of these limitations, little is known about the prevalence of autoimmunity outside wealthy regions and clinical settings. AIMS The present paper discusses why AIID are of critical importance in human biology research and why more work needs to be devoted to validating, testing, and utilizing methods for detecting autoantibodies and other biomarkers related to autoimmunity in field-friendly, minimally invasively-collected samples. This paper reviews some of the methods used to diagnose AIIDs in clinical settings, and highlights methods that have been used in studies within human biology and related fields, emphasizing the invasiveness of specific methods and their feasibility in remote field settings. DISCUSSION AND CONCLUSIONS Risk for AIID is affected by several reproductive, dietary, environmental, and genetic factors. Human biologists have unique perspectives that they can bring to autoimmunity research, and more population-based studies on autoimmunity are needed within these and related fields.
Collapse
Affiliation(s)
- Tara J Cepon-Robins
- Department of Anthropology, University of Colorado Colorado Springs, Colorado Springs, Colorado, USA
| |
Collapse
|
48
|
Li Y, Zhou Y, Zhao M, Zou J, Zhu Y, Yuan X, Liu Q, Cai H, Chu CQ, Liu Y. Differential Profile of Plasma Circular RNAs in Type 1 Diabetes Mellitus. Diabetes Metab J 2020; 44:854-865. [PMID: 32662258 PMCID: PMC7801755 DOI: 10.4093/dmj.2019.0151] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/30/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND No currently available biomarkers or treatment regimens fully meet therapeutic needs of type 1 diabetes mellitus (T1DM). Circular RNA (circRNA) is a recently identified class of stable noncoding RNA that have been documented as potential biomarkers for various diseases. Our objective was to identify and analyze plasma circRNAs altered in T1DM. METHODS We used microarray to screen differentially expressed plasma circRNAs in patients with new onset T1DM (n=3) and age-/gender-matched healthy controls (n=3). Then, we selected six candidates with highest fold-change and validated them by quantitative real-time polymerase chain reaction in independent human cohort samples (n=12). Bioinformatic tools were adopted to predict putative microRNAs (miRNAs) sponged by these validated circRNAs and their downstream messenger RNAs (mRNAs). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to gain further insights into T1DM pathogenesis. RESULTS We identified 68 differentially expressed circRNAs, with 61 and seven being up- and downregulated respectively. Four of the six selected candidates were successfully validated. Curations of their predicted interacting miRNAs revealed critical roles in inflammation and pathogenesis of autoimmune disorders. Functional relations were visualized by a circRNA-miRNA-mRNA network. GO and KEGG analyses identified multiple inflammation-related processes that could be potentially associated with T1DM pathogenesis, including cytokine-cytokine receptor interaction, inflammatory mediator regulation of transient receptor potential channels and leukocyte activation involved in immune response. CONCLUSION Our study report, for the first time, a profile of differentially expressed plasma circRNAs in new onset T1DM. Further in silico annotations and bioinformatics analyses supported future application of circRNAs as novel biomarkers of T1DM.
Collapse
Affiliation(s)
- Yangyang Li
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun, China
| | - Ying Zhou
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Minghui Zhao
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun, China
| | - Jing Zou
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Yuxiao Zhu
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Xuewen Yuan
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Qianqi Liu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hanqing Cai
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun, China
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University School of Medicine, Portland, OR, USA
- Section of Rheumatology, VA Portland Health Care System, Portland, OR, USA
| | - Yu Liu
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
49
|
Grohová A, Dáňová K, Adkins I, Šumník Z, Petruželková L, Obermannová B, Koloušková S, Špíšek R, Palová-Jelínková L. Myeloid - derived suppressor cells in Type 1 diabetes are an expanded population exhibiting diverse T-cell suppressor mechanisms. PLoS One 2020; 15:e0242092. [PMID: 33206686 PMCID: PMC7673497 DOI: 10.1371/journal.pone.0242092] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/26/2020] [Indexed: 12/11/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) represent a heterogeneous group of immature myeloid cells with immunoregulatory function in cancer and autoimmune diseases. In humans, two subsets of MDSC were determined based on the characteristic surface markers, monocytic MDSC (M-MDSC) and granulocytic MDSC (G-MDSC). Expansion of MDSC has been reported in some murine models and patients with autoimmune diseases and their immune-suppressive properties were characterized. However, the exact role of MDSC in the pathogenesis of autoimmune diseases is more complex and/or controversial. In type 1 diabetes mellitus (T1D), the increased frequency of MDSC was found in the blood of T1D patients but their suppressor capacity was diminished. In our study, we assessed the role of M-MDSC in the pathogenesis of T1D and showed for the first time the increased frequency of M-MDSC not only in the blood of T1D patients but also in their at-risk relatives compared to healthy donors. T1D patients with inadequate long term metabolic control showed an elevation of M-MDSC compared to patients with better disease control. Furthermore, we described the positive correlation between the percentage of M-MDSC and Th17 cells and IFN-γ producing T cells in T1D patients and their at-risk relatives. Finally, we found that the ability of M-MDSC to suppress autologous T cells is efficient only at the high MDSC: T cells ratio and dependent on cell-cell-contact and TGF-β production. Our data show that the engagement of MDSC in the pathogenesis of T1D is evident, yet not entirely explored and more experiments are required to clarify whether MDSC are beneficial or harmful in T1D.
Collapse
Affiliation(s)
- Anna Grohová
- Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Klára Dáňová
- Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,SOTIO a.s., Prague, Czech Republic
| | - Irena Adkins
- Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,SOTIO a.s., Prague, Czech Republic
| | - Zdeněk Šumník
- Department of Pediatrics, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Lenka Petruželková
- Department of Pediatrics, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Barbora Obermannová
- Department of Pediatrics, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Stanislava Koloušková
- Department of Pediatrics, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Radek Špíšek
- Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,SOTIO a.s., Prague, Czech Republic
| | - Lenka Palová-Jelínková
- Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,SOTIO a.s., Prague, Czech Republic
| |
Collapse
|
50
|
Bolandi V, Azghadi SK, Shahami M, Fereidouni M. Prevalence of IA-2 antibody in patients suffering from diabetes and their first-degree relatives. Int J Diabetes Dev Ctries 2020. [DOI: 10.1007/s13410-020-00882-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|