1
|
Guan W, Zhang L. Applications and prospects of biomaterials in diabetes management. Front Bioeng Biotechnol 2025; 13:1547343. [PMID: 40124248 PMCID: PMC11926158 DOI: 10.3389/fbioe.2025.1547343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/30/2025] [Indexed: 03/25/2025] Open
Abstract
Diabetes is a widespread metabolic disorder that presents considerable challenges in its management. Recent advancements in biomaterial research have shed light on innovative approaches for the treatment of diabetes. This review examines the role of biomaterials in diabetes diagnosis and treatment, as well as their application in managing diabetic wounds. By evaluating recent research developments alongside future obstacles, the review highlights the promising potential of biomaterials in diabetes care, underscoring their importance in enhancing patient outcomes and refining treatment methodologies.
Collapse
Affiliation(s)
- Wenhe Guan
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liang Zhang
- Department of Human Anatomy, School of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning, China
| |
Collapse
|
2
|
Korpijaakko CA, Eriksson JG, Kautiainen H, Klemetti MM, Laine MK. Increased purchases of prescription medicines in offspring of women with type 1 diabetes: a Finnish register-based cohort study between 1995 and 2018. Ann Med 2024; 56:2412283. [PMID: 39434552 PMCID: PMC11497570 DOI: 10.1080/07853890.2024.2412283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 05/12/2024] [Accepted: 06/05/2024] [Indexed: 10/23/2024] Open
Abstract
OBJECTIVE This study aimed to assess whether in utero exposure to hyperglycemia influences prescription medicine purchases in the offspring of women with type 1 diabetes (exposed offspring). PATIENTS/MATERIALS AND METHODS We identified all singleton exposed offspring born in the hospital district of Helsinki and Uusimaa, Finland, between 1988 and 2011 from the Finnish Medical Birth Register, maintained by the Finnish Institute for Health and Welfare. For each exposed offspring, we obtained five age- and province-matched offspring of women without diabetes (reference offspring), from the Finnish Medical Birth Register. By combining data from three national registers, this longitudinal cohort study assessed prescription medicine purchases in exposed offspring (n = 1,725) and reference offspring (n = 8,755) from seven to thirty years of age. Prescription medicine purchases were grouped according to the Anatomical Therapeutic Chemical (ATC) classification system. RESULTS Between 1995 and 2018, a total of 211,490 prescription medicines were purchased. After a median follow-up of 10.9 (interquartile range 4.9,17.6) years, we observed higher incidence risk ratios (IRR) of prescription medicine purchases for several ATC main groups in exposed offspring compared to reference offspring, with the highest IRR of 4.06 (95% CI: 2.78 to 5.94) for medicines affecting metabolism (e.g. diabetes medicines). CONCLUSION Our findings suggest that exposed offspring purchase more reimbursable prescription medicines than reference offspring from age seven to thirty years. More research is needed to examine the effects of intrauterine exposure to hyperglycemia on long-term health in offspring.
Collapse
Affiliation(s)
- Cedric A. Korpijaakko
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Johan G. Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Yong Loo Lin School of Medicine, Human Potential Translational Research Programme and Department of Obstetrics and Gynecology, National University Singapore, Singapore, Singapore
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hannu Kautiainen
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Primary Health Care Unit, Kuopio University Hospital, Kuopio, Finland
| | - Miira M. Klemetti
- Department of Obstetrics and Gynecology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Merja K. Laine
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| |
Collapse
|
3
|
Mănescu M, Mănescu IB, Grama A. A Review of Stage 0 Biomarkers in Type 1 Diabetes: The Holy Grail of Early Detection and Prevention? J Pers Med 2024; 14:878. [PMID: 39202069 PMCID: PMC11355657 DOI: 10.3390/jpm14080878] [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: 08/04/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Type 1 diabetes mellitus (T1D) is an incurable autoimmune disease characterized by the destruction of pancreatic islet cells, resulting in lifelong dependency on insulin treatment. There is an abundance of review articles addressing the prediction of T1D; however, most focus on the presymptomatic phases, specifically stages 1 and 2. These stages occur after seroconversion, where therapeutic interventions primarily aim to delay the onset of T1D rather than prevent it. This raises a critical question: what happens before stage 1 in individuals who will eventually develop T1D? Is there a "stage 0" of the disease, and if so, how can we detect it to increase our chances of truly preventing T1D? In pursuit of answers to these questions, this narrative review aimed to highlight recent research in the field of early detection and prediction of T1D, specifically focusing on biomarkers that can predict T1D before the onset of islet autoimmunity. Here, we have compiled influential research from the fields of epigenetics, omics, and microbiota. These studies have identified candidate biomarkers capable of predicting seroconversion from very early stages to several months prior, suggesting that the prophylactic window begins at birth. As the therapeutic landscape evolves from treatment to delay, and ideally from delay to prevention, it is crucial to both identify and validate such "stage 0" biomarkers predictive of islet autoimmunity. In the era of precision medicine, this knowledge will enable early intervention with the potential for delaying, modifying, or completely preventing autoimmunity and T1D in at-risk children.
Collapse
Affiliation(s)
- Măriuca Mănescu
- Department of Pediatrics, Emergency County Clinical Hospital of Targu Mures, 50 Gheorghe Marinescu, 540136 Targu Mures, Romania;
| | - Ion Bogdan Mănescu
- Department of Laboratory Medicine, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 38 Gheorghe Marinescu, 540142 Targu Mures, Romania;
| | - Alina Grama
- Department of Pediatrics, Emergency County Clinical Hospital of Targu Mures, 50 Gheorghe Marinescu, 540136 Targu Mures, Romania;
- Department of Pediatrics, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 38 Gheorghe Marinescu, 540142 Targu Mures, Romania
| |
Collapse
|
4
|
Urrutia I, Martinez R, Calvo B, Marcelo I, Saso-Jimenez L, Martinez de Lapiscina I, Bilbao JR, Castano L, Rica I. Risk for progression to type 1 diabetes in first-degree relatives under 50 years of age. Front Endocrinol (Lausanne) 2024; 15:1411686. [PMID: 39188918 PMCID: PMC11345149 DOI: 10.3389/fendo.2024.1411686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024] Open
Abstract
Introduction The detection of pancreatic autoantibodies in first-degree relatives of patients with type 1 diabetes (T1D) is considered a risk factor for disease. Novel available immunotherapies to delay T1D progression highlight the importance of identifying individuals at risk who might benefit from emerging treatments. The objective was to assess the autoimmunity in first-degree relatives of patients with T1D, estimate the time from autoimmunity detection to the onset of clinical diabetes, and identify the associated risk factors. Methods Retrospective multicenter study of 3,015 first-degree relatives of patients with T1D recruited between 1992 and 2018. Pancreatic autoantibodies (IAA, GADA, IA2A, and ZnT8A) were determined by radioimmunoassay, starting the analyses at diagnosis of the proband. All those with positive autoimmunity and normal fasting blood glucose without clinical symptoms of diabetes were followed up in the study. The progression rate to T1D was assessed according to sex, relationship with the proband, age at autoimmunity detection, type/number of autoantibodies, and HLA-DRB1 genotype. Cox proportional-hazard models and Kaplan-Meier survival plots were used for statistical analyses. Results Among the relatives, 21 progenitors [43.7 years (IQR: 38.1-47.7)] and 27 siblings [7.6 years (IQR: 5.8-16.1)] had positive autoantibodies. Of these, 54.2% (95% CI: 39.2%-68.6%) developed T1D (age at autoimmunity detection 11 months to 39 years) in a median of 5 years (IQR: 3.6-8.7; ranged from 0.9 to 22.6 years). Risk factors associated with faster progression to T1D were multiple autoimmunity and <20 years at autoimmunity detection. Younger relatives (<20 years) with multiple autoantibodies had a 5-year cumulative risk of developing diabetes of 52.9% (95% CI: 22.1%-71.6%) and a 20-year risk of 91.2% (95% CI: 50.5%-98.4%). The 20-year risk decreased to 59.9% (95% CI: 21.9%-79.5%) if only one risk factor was met and to 35.7% (95% CI: 0.0%-66.2%) if the relative was older than 20 years with one autoantibody. Conclusions In first-degree relatives with autoimmunity, the time to progression to T1D is faster in children and adolescents with multiple autoantibodies. Young adults are also at risk, which supports their consideration in screening strategies for people at risk of developing T1D.
Collapse
Affiliation(s)
- Ines Urrutia
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Rosa Martinez
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Begona Calvo
- Biobizkaia Health Research Institute, Barakaldo, Spain
- Department of Medical Oncology, Cruces University Hospital, Barakaldo, Spain
| | - Irene Marcelo
- Hospital de Mataró - Consorci Sanitari del Maresme, Barcelona, Spain
| | - Laura Saso-Jimenez
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Idoia Martinez de Lapiscina
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Jose Ramon Bilbao
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Luis Castano
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
| | - Itxaso Rica
- Biobizkaia Health Research Institute, Barakaldo, Spain
- UPV/EHU, CIBERDEM, CIBERER, Endo-ERN, Barakaldo, Spain
- Pediatric Endocrinology Unit, Cruces University Hospital, Barakaldo, Spain
| |
Collapse
|
5
|
Robino A, Bevilacqua E, Aldegheri L, Conti A, Bazzo V, Tornese G, Catamo E. Next-generation sequencing reveals additional HLA class I and class II alleles associated with type 1 diabetes and age at onset. Front Immunol 2024; 15:1427349. [PMID: 39185409 PMCID: PMC11341356 DOI: 10.3389/fimmu.2024.1427349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
Introduction Type 1 diabetes is an autoimmune disease with an significant genetic component, played mainly by the HLA class II genes. Although evidence on the role of HLA class I genes in developing type 1 diabetes and its onset have emerged, current HLA screening is limited to determining DR3 and DR4 haplotypes. This study aimed to investigate the role of HLA genes on type 1 diabetes risk and age of onset by extensive typing. Methods This study included 115 children and young adults with type 1 diabetes for whom typing of HLA-A, -B, -C, -DRB1, -DRB3/4/5, -DQA1, -DQB1, -DPA1 and -DPB1 genes was conducted using Next Generation Sequencing. Results We observed that 13% of type 1 diabetes subjects had non-classical HLA haplotypes that predispose to diabetes. We also found that compared to type 1 diabetes subjects with classical HLA haplotypes, non-classical HLA subjects had a significantly higher frequency of HLA-B*39:06:02 (p-value=0.01) and HLA-C*07:02:01 (p-value=0.03) alleles, known to be involved in activating the immune response. Non-classical HLA subjects also presented peculiar clinical features compared to classical HLA subjects, such as multiple diabetic antibodies and the absence of other autoimmune diseases (i.e., coeliac disease and thyroiditis). We also observed that subjects with early onset had a higher frequency of DQ2/DQ8 genotype than late-onset individuals. Moreover, subjects with late-onset had a higher frequency of alleles HLA-B*27 (p-value=0.003), HLA-C*01:02:01 (p-value=0.027) and C*02:02:02 (p-value=0.01), known to be associated with increased protection against viral infections. Discussion This study reveals a broader involvement of the HLA locus in the development and onset of type 1 diabetes, providing insights into new possible disease prevention and management strategies.
Collapse
Affiliation(s)
- Antonietta Robino
- Institute for Maternal and Child Health – IRCCS Burlo Garofolo, Trieste, Italy
| | - Elena Bevilacqua
- Transfusion Medicine Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Luana Aldegheri
- Institute for Maternal and Child Health – IRCCS Burlo Garofolo, Trieste, Italy
| | - Andrea Conti
- Institute for Maternal and Child Health – IRCCS Burlo Garofolo, Trieste, Italy
| | - Valentina Bazzo
- Transfusion Medicine Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Gianluca Tornese
- Institute for Maternal and Child Health – IRCCS Burlo Garofolo, Trieste, Italy
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Eulalia Catamo
- Institute for Maternal and Child Health – IRCCS Burlo Garofolo, Trieste, Italy
| |
Collapse
|
6
|
Bawatneh A, Darwish A, Eideh H, Darwish HM. Identification of gene mutations associated with type 1 diabetes by next-generation sequencing in affected Palestinian families. Front Genet 2024; 14:1292073. [PMID: 38274107 PMCID: PMC10808782 DOI: 10.3389/fgene.2023.1292073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/04/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction: Diabetes Mellitus is a group of metabolic disorders characterized by hyperglycemia secondary to insulin resistance or deficiency. It is considered a major health problem worldwide. T1DM is a result of a combination of genetics, epigenetics, and environmental factors. Several genes have been associated with T1DM, including HLA, INS, CTLA4, and PTPN22. However, none of these findings have been based on linkage analysis because it is rare to find families with several diabetic individuals. Two Palestinian families with several afflicted members with variations in the mode of inheritance were identified and selected for this study. This study aimed to identify the putative causative gene(s) responsible for T1DM development in these families to improve our understanding of the molecular genetics of the disease. Methods: One afflicted member from each family was selected for Whole-Exome Sequencing. Data were mapped to the reference of the human genome, and the resulting VCF file data were filtered. The variants with the highest phenotype correlation score were checked by Sanger sequencing for all family members. The confirmed variants were analyzed in silico by bioinformatics tools. Results: In one family, the IGF1R p.V579F variant, which follows autosomal dominant inheritance, was confirmed and segregated in the family. In another family, the NEUROD1 p.P197H variant, which follows autosomal recessive inheritance, was positively confirmed and segregated. Conclusion: IGF1R p.V579F and NEUROD1 p.P197H variants were associated with T1DM development in the two inflicted families. Further analysis and functional assays will be performed, including the generation of mutant model cell systems, to unravel their specific molecular mechanism in the disease development.
Collapse
Affiliation(s)
- Abrar Bawatneh
- Molecular Genetics and Genetics Toxicology Program, Faculty of Graduate Studies, Arab American University, Jenin, Palestine
| | - Alaa Darwish
- Faculty of Health Professions, AlQuds University, Jerusalem, Palestine
| | | | - Hisham M. Darwish
- Molecular Genetics and Genetics Toxicology Program, Faculty of Graduate Studies, Arab American University, Jenin, Palestine
- Faculty of Allied Medical Sciences, Arab American University, Jenin, Palestine
| |
Collapse
|
7
|
Niazi SK. Anti-Idiotypic mRNA Vaccine to Treat Autoimmune Disorders. Vaccines (Basel) 2023; 12:9. [PMID: 38276668 PMCID: PMC10819008 DOI: 10.3390/vaccines12010009] [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: 10/02/2023] [Revised: 11/01/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
The 80+ existing autoimmune disorders (ADs) affect billions with little prevention or treatment options, except for temporary symptomatic management, leading to enormous human suffering and a monumental financial burden. The autoantibodies formed in most ADs have been identified, allowing the development of novel anti-idiotypic antibodies to mute the autoantibodies using vaccines. Nucleoside vaccines have been successfully tested as antigen-specific immunotherapies (ASI), with mRNA technology offering multi-epitope targeting to mute multiple autoantibodies. This paper proposes using mRNA technology to produce anti-idiotypic antibodies with broad effectiveness in preventing and treating them. This paper delves into the state-of-the-art mRNA design strategies used to develop novel ASIs by selecting appropriate T cell and B cell epitopes to generate anti-idiotypic antibodies. The low cost and fast development of mRNA vaccines make this technology the most affordable for the global control of ADs.
Collapse
Affiliation(s)
- Sarfaraz K Niazi
- College of Pharmacy, University of Illinois, Chicago, IL 60012, USA
| |
Collapse
|
8
|
Urrutia I, Martínez R, Calvo B, Saso-Jiménez L, González P, Fernández-Rubio E, Martín-Nieto A, Aguayo A, Rica I, Gaztambide S, Castano L. Autoimmune Diabetes From Childhood to Adulthood: The Role of Pancreatic Autoantibodies and HLA-DRB1 Genotype. J Clin Endocrinol Metab 2023; 108:e1341-e1346. [PMID: 37207452 DOI: 10.1210/clinem/dgad277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/21/2023]
Abstract
CONTEXT Autoimmune diabetes can develop at any age, but unlike early-onset diabetes, adult onset is less well documented. We aimed to compare, over a wide age range, the most reliable predictive biomarkers for this pathology: pancreatic-autoantibodies and HLA-DRB1 genotype. METHODS A retrospective study of 802 patients with diabetes (aged 11 months to 66 years) was conducted. Pancreatic autoantibodies at diagnosis: insulin autoantibodies (IAA), glutamate decarboxylase autoantibodies (GADA), islet tyrosine phosphatase 2 autoantibodies (IA2A), and zinc transporter-8 autoantibodies (ZnT8A) and HLA-DRB1 genotype were analyzed. RESULTS Compared with early-onset patients, adults had a lower frequency of multiple autoantibodies, with GADA being the most common. At early onset, IAA was the most frequent in those younger than 6 years and correlated inversely with age; GADA and ZnT8A correlated directly and IA2A remained stable.The absence of HLA-DRB1 risk genotype was associated with higher age at diabetes onset (27.5 years; interquartile range [IQR], 14.3-35.7), whereas the high-risk HLA-DR3/DR4 was significantly more common at lower age (11.9 years; IQR, 7.1-21.6). ZnT8A was associated with DR4/non-DR3 (odds ratio [OR], 1.91; 95% CI, 1.15-3.17), GADA with DR3/non-DR4 (OR, 2.97; 95% CI, 1.55-5.71), and IA2A with DR4/non-DR3 and DR3/DR4 (OR, 3.89; 95% CI, 2.28-6.64, and OR, 3.08; 95% CI, 1.83-5.18, respectively). No association of IAA with HLA-DRB1 was found. CONCLUSION Autoimmunity and HLA-DRB1 genotype are age-dependent biomarkers. Adult-onset autoimmune diabetes is associated with lower genetic risk and lower immune response to pancreatic islet cells compared with early-onset diabetes.
Collapse
Affiliation(s)
- Inés Urrutia
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
| | - Rosa Martínez
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
| | - Begona Calvo
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- Department of Medical Oncology, Hospital Universitario Cruces, 48903 Barakaldo, Spain
| | - Laura Saso-Jiménez
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
| | - Pedro González
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- Department of Endocrinology and Nutrition, Hospital Universitario Cruces, 48903 Barakaldo, Spain
| | - Elsa Fernández-Rubio
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- Department of Endocrinology and Nutrition, Hospital Universitario Cruces, 48903 Barakaldo, Spain
| | - Alicia Martín-Nieto
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- Department of Endocrinology and Nutrition, Hospital Universitario Cruces, 48903 Barakaldo, Spain
| | - Anibal Aguayo
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
| | - Itxaso Rica
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
- Department of Pediatric Endocrinology, Hospital Universitario Cruces, 48903 Barakaldo, Spain
| | - Sonia Gaztambide
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
- Department of Endocrinology and Nutrition, Hospital Universitario Cruces, 48903 Barakaldo, Spain
| | - Luis Castano
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
| |
Collapse
|
9
|
Zargari I, Adar A, Morag I, Pinhas-Hamiel O, Eyal O, Keidar R, Loewenthal N, Levy M, Dally-Gottfried O, Landau Z, Levy-Khademi F, Eventov-Friedman S, Zangen D, Youngster I, Rachmiel M. Early exposures and inherent factors in premature newborns are associated with type 1 diabetes. Pediatr Res 2023; 94:1516-1522. [PMID: 35488031 DOI: 10.1038/s41390-022-02069-w] [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: 10/02/2020] [Revised: 02/08/2022] [Accepted: 02/27/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Pathophysiology of type 1 diabetes (T1D) involves immune responses that may be associated with early exposure to environmental factors among preterm newborns. The aim of this work was to evaluate for association between T1D and maternal, nutritional, and medical exposures during the neonatal period among premature newborns. METHODS This is a multicenter, matched case-control study. Preterm newborns, who developed T1D before 18 years, were matched by sex, gestational age (GA), birth date, and medical center of birth with newborns who did not develop TID. Data included maternal medical history, birth weight (BW), length of hospitalization, enteral and parenteral medications, fluid administration, and feeding modalities during hospitalization. RESULTS Fifty-two patients with T1D, 26 males, median age at T1D diagnosis 8.17 years (5.92-9.77), median GA 34 weeks (33-m36), and 132 matched controls, were included. Multivariate-conditional-regression demonstrated a significant association between T1D and any maternal illness (23.1% vs. 9.1%, OR = 4.99 (1.69-14.72), p = 0.004), higher BW-SDS (0.07 ± 0.95 vs. -0.27 ± 0.97, OR = 2.03 (1.19-3.49), p = 0.01), longer duration of glucose infusion (3 (1-5) days vs. 2 (0-4), OR = 1.23 (1.03-1.46), p = 0.02), and antibiotic therapy beyond the first week of life (19.2% vs. 6.9%, OR = 5.22 (1.32-20.70), p = 0.019). Antibiotic treatment during the first week of life was negatively associated with T1D (51.9% vs. 67.2%, OR 0.31 (0.11-0.88), p = 0.027). CONCLUSIONS A novel association was demonstrated between the development of T1D and early interventions and exposures among preterm newborns. IMPACT Type 1 diabetes mellitus during childhood may be associated with early exposures during the neonatal period, in addition to known maternal and neonatal metabolic parameters. Early exposure to intravenous antibiotics, differing between the first week of life and later, and longer parenteral glucose administration to preterm newborns were associated with childhood type 1 diabetes. This is in addition to familiar maternal risk factors. Future prospective studies should examine the microbial changes and immune system characteristics of preterm and term neonates exposed to parenteral antibiotics and glucose treatment, in order to validate our exploratory findings.
Collapse
Affiliation(s)
- Iren Zargari
- Pediatric Endocrinology Unit, Division of Pediatrics, Shamir Medical Center (Assaf Harofeh), Zerifin, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adi Adar
- Pediatric Endocrinology Unit, Division of Pediatrics, Shamir Medical Center (Assaf Harofeh), Zerifin, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Iris Morag
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Neonatal Intensive Care Unit, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel
| | - Orit Pinhas-Hamiel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Endocrinology and Diabetes Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel
| | - Ori Eyal
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Endocrinology and Diabetes Unit, Dana-Dwek Children Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Rimona Keidar
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Neonatal Intensive Care Unit, Shamir Medical Center (Assaf Harofeh), Zerifin, Israel
| | - Neta Loewenthal
- Pediatric Endocrinology and Metabolic Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Milana Levy
- Pediatric Diabetes and Obesity Clinic, Ruth Rappaport Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Orna Dally-Gottfried
- The Center for Juvenile Diabetes and Pediatric Endocrinology and Pediatric Outpatient Clinics, Rebecca Ziv Hospital, affiliated to The School of Medicine, Bar Ilan University, Safed, Israel
| | - Zohar Landau
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Endocrinology and Diabetes Unit, E. Wolfson Medical Center, Holon, Israel
| | - Floris Levy-Khademi
- The School of Medicine in the Hebrew University of Jerusalem, Jerusalem, Israel
- Division of Pediatric Endocrinology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Smadar Eventov-Friedman
- The School of Medicine in the Hebrew University of Jerusalem, Jerusalem, Israel
- Neonatal Intensive Care Unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - David Zangen
- The School of Medicine in the Hebrew University of Jerusalem, Jerusalem, Israel
- Division of Pediatric Endocrinology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ilan Youngster
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Pediatric Infectious Diseases Unit, Shamir Medical Center (Assaf Harofeh), Zerifin, Israel
| | - Marianna Rachmiel
- Pediatric Endocrinology Unit, Division of Pediatrics, Shamir Medical Center (Assaf Harofeh), Zerifin, Israel.
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
10
|
Mousa M, Albarguthi S, Albreiki M, Farooq Z, Sajid S, El Hajj Chehadeh S, ElBait GD, Tay G, Deeb AA, Alsafar H. Whole-Exome Sequencing in Family Trios Reveals De Novo Mutations Associated with Type 1 Diabetes Mellitus. BIOLOGY 2023; 12:biology12030413. [PMID: 36979105 PMCID: PMC10044903 DOI: 10.3390/biology12030413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/16/2023] [Accepted: 02/23/2023] [Indexed: 03/10/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic autoimmune disease characterized by insulin deficiency and loss of pancreatic islet β-cells. The objective of this study is to identify de novo mutations in 13 trios from singleton families that contribute to the genetic basis of T1DM through the application of whole-exome sequencing (WES). Of the 13 families sampled for this project, 12 had de novo variants, with Family 7 having the highest number (nine) of variants linked to T1DM/autoimmune pathways, whilst Family 4 did not have any variants past the filtering steps. There were 10 variants of 7 genes reportedly associated with T1DM (MST1; TDG; TYRO3; IFIHI; GLIS3; VEGFA; TYK2). There were 20 variants of 13 genes that were linked to endocrine, metabolic, or autoimmune diseases. Our findings demonstrate that trio-based WES is a powerful approach for identifying new candidate genes for the pathogenesis of T1D. Genotyping and functional annotation of the discovered de novo variants in a large cohort is recommended to ascertain their association with disease pathogenesis.
Collapse
Affiliation(s)
- Mira Mousa
- Center of Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Sara Albarguthi
- Center of Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Mohammed Albreiki
- Center of Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Zenab Farooq
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Sameeha Sajid
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
| | - Sarah El Hajj Chehadeh
- Center of Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Gihan Daw ElBait
- Center of Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Guan Tay
- Center of Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
| | - Asma Al Deeb
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi 127788, United Arab Emirates
- Department of Endocrinology, Mafraq Hospital, Abu Dhabi 127788, United Arab Emirates
| | - Habiba Alsafar
- Center of Biotechnology, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi 127788, United Arab Emirates
- Correspondence:
| |
Collapse
|
11
|
Identification of novel differentially expressed genes in type 1 diabetes mellitus complications using transcriptomic profiling of UAE patients: a multicenter study. Sci Rep 2022; 12:16316. [PMID: 36175575 PMCID: PMC9523055 DOI: 10.1038/s41598-022-18997-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 08/23/2022] [Indexed: 12/01/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic metabolic disorder that mainly affects children and young adults. It is associated with debilitating and long-life complications. Therefore, understanding the factors that lead to the onset and development of these complications is crucial. To our knowledge this is the first study that attempts to identify the common differentially expressed genes (DEGs) in T1DM complications using whole transcriptomic profiling in United Arab Emirates (UAE) patients. The present multicenter study was conducted in different hospitals in UAE including University Hospital Sharjah, Dubai Hospital and Rashid Hospital. A total of fifty-eight Emirati participants aged above 18 years and with a BMI < 25 kg/m2 were recruited and forty-five of these participants had a confirmed diagnosis of T1DM. Five groups of complications associated with the latter were identified including hyperlipidemia, neuropathy, ketoacidosis, hypothyroidism and polycystic ovary syndrome (PCOS). A comprehensive whole transcriptomic analysis using NGS was conducted. The outcomes of the study revealed the common DEGs between T1DM without complications and T1DM with different complications. The results revealed seven common candidate DEGs, SPINK9, TRDN, PVRL4, MYO3A, PDLIM1, KIAA1614 and GRP were upregulated in T1DM complications with significant increase in expression of SPINK9 (Fold change: 5.28, 3.79, 5.20, 3.79, 5.20) and MYO3A (Fold change: 4.14, 6.11, 2.60, 4.33, 4.49) in hyperlipidemia, neuropathy, ketoacidosis, hypothyroidism and PCOS, respectively. In addition, functional pathways of ion transport, mineral absorption and cytosolic calcium concentration were involved in regulation of candidate upregulated genes related to neuropathy, ketoacidosis and PCOS, respectively. The findings of this study represent a novel reference warranting further studies to shed light on the causative genetic factors that are involved in the onset and development of T1DM complications.
Collapse
|
12
|
Rojas M, Heuer LS, Zhang W, Chen YG, Ridgway WM. The long and winding road: From mouse linkage studies to a novel human therapeutic pathway in type 1 diabetes. Front Immunol 2022; 13:918837. [PMID: 35935980 PMCID: PMC9353112 DOI: 10.3389/fimmu.2022.918837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Autoimmunity involves a loss of immune tolerance to self-proteins due to a combination of genetic susceptibility and environmental provocation, which generates autoreactive T and B cells. Genetic susceptibility affects lymphocyte autoreactivity at the level of central tolerance (e.g., defective, or incomplete MHC-mediated negative selection of self-reactive T cells) and peripheral tolerance (e.g., failure of mechanisms to control circulating self-reactive T cells). T regulatory cell (Treg) mediated suppression is essential for controlling peripheral autoreactive T cells. Understanding the genetic control of Treg development and function and Treg interaction with T effector and other immune cells is thus a key goal of autoimmunity research. Herein, we will review immunogenetic control of tolerance in one of the classic models of autoimmunity, the non-obese diabetic (NOD) mouse model of autoimmune Type 1 diabetes (T1D). We review the long (and still evolving) elucidation of how one susceptibility gene, Cd137, (identified originally via linkage studies) affects both the immune response and its regulation in a highly complex fashion. The CD137 (present in both membrane and soluble forms) and the CD137 ligand (CD137L) both signal into a variety of immune cells (bi-directional signaling). The overall outcome of these multitudinous effects (either tolerance or autoimmunity) depends upon the balance between the regulatory signals (predominantly mediated by soluble CD137 via the CD137L pathway) and the effector signals (mediated by both membrane-bound CD137 and CD137L). This immune balance/homeostasis can be decisively affected by genetic (susceptibility vs. resistant alleles) and environmental factors (stimulation of soluble CD137 production). The discovery of the homeostatic immune effect of soluble CD137 on the CD137-CD137L system makes it a promising candidate for immunotherapy to restore tolerance in autoimmune diseases.
Collapse
Affiliation(s)
- Manuel Rojas
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
- School of Medicine and Health Sciences, Doctoral Program in Biological and Biomedical Sciences, Universidad del Rosario, Bogota, Colombia
| | - Luke S. Heuer
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Weici Zhang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
| | - Yi-Guang Chen
- The Max McGee Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Wisconsin, Milwaukee, WI, United States
- Division of Endocrinology, Department of Pediatrics, The Medical College of Wisconsin, Milwaukee, WI, United States
| | - William M. Ridgway
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
- *Correspondence: William M. Ridgway,
| |
Collapse
|
13
|
Massignam ET, Dieter C, Assmann TS, Duarte GCK, Bauer AC, Canani LH, Crispim D. The rs705708 A allele of the ERBB3 gene is associated with lower prevalence of diabetic retinopathy and arterial hypertension and with improved renal function in type 1 diabetic patients. Microvasc Res 2022; 143:104378. [PMID: 35594935 DOI: 10.1016/j.mvr.2022.104378] [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: 03/07/2022] [Revised: 04/21/2022] [Accepted: 05/11/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The Erb-b2 receptor tyrosine kinase 3 (ERBB3) is involved in autoimmune processes related to type 1 diabetes mellitus (T1DM) pathogenesis. Accordingly, some studies have suggested that single nucleotide polymorphisms (SNPs) in the ERBB3 gene confer risk for T1DM. Proliferation-associated protein 2G4 (PA2G4) is another candidate gene for this disease because it regulates cell proliferation and adaptive immunity. Moreover, PA2G4 regulates ERBB3. To date, no study has evaluated the association of PA2G4 SNPs and T1DM. AIM To evaluate the association of ERBB3 rs705708 (G/A) and PA2G4 rs773120 (C/T) SNPs with T1DM and its clinical and laboratory characteristics. METHODS This case-control study included 976 white subjects from Southern Brazil, categorized into 501 cases with T1DM and 475 non-diabetic controls. The ERBB3 and PA2G4 SNPs were genotyped by allelic discrimination-real-time PCR. RESULTS ERBB3 rs705708 and PA2G4 rs773120 SNPs were not associated with T1DM considering different inheritance models and also when controlling for covariables. However, T1DM patients carrying the ERBB3 rs705708 A allele developed T1DM at an earlier age vs. G/G patients. Interestingly, in the T1DM group, the rs705708 A allele was associated with lower prevalence of diabetic retinopathy and arterial hypertension as well as with improved renal function (higher estimated glomerular filtration rate and lower urinary albumin excretion levels) compared to G/G patients. CONCLUSIONS Although no association was observed between the ERBB3 rs705708 and PA2G4 rs773120 SNPs and T1DM, the rs705708 A allele was associated, for the first time in literature, with lower prevalence of diabetic retinopathy and arterial hypertension. Additionally, this SNP was associated with improved renal function.
Collapse
Affiliation(s)
- Eloísa Toscan Massignam
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, Rio Grande do Sul, Brazil
| | - Cristine Dieter
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, Rio Grande do Sul, Brazil
| | - Taís Silveira Assmann
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, Rio Grande do Sul, Brazil
| | - Guilherme Coutinho Kullmann Duarte
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, Rio Grande do Sul, Brazil
| | - Andrea Carla Bauer
- Nephrology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Luis Henrique Canani
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, Rio Grande do Sul, Brazil
| | - Daisy Crispim
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
14
|
Kim J, Jensen A, Ko S, Raghavan S, Phillips LS, Hung A, Sun Y, Zhou H, Reaven P, Zhou JJ. Systematic Heritability and Heritability Enrichment Analysis for Diabetes Complications in UK Biobank and ACCORD Studies. Diabetes 2022; 71:1137-1148. [PMID: 35133398 PMCID: PMC9044130 DOI: 10.2337/db21-0839] [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: 09/16/2021] [Accepted: 02/02/2022] [Indexed: 11/13/2022]
Abstract
Diabetes-related complications reflect longstanding damage to small and large vessels throughout the body. In addition to the duration of diabetes and poor glycemic control, genetic factors are important contributors to the variability in the development of vascular complications. Early heritability studies found strong familial clustering of both macrovascular and microvascular complications. However, they were limited by small sample sizes and large phenotypic heterogeneity, leading to less accurate estimates. We take advantage of two independent studies-UK Biobank and the Action to Control Cardiovascular Risk in Diabetes trial-to survey the single nucleotide polymorphism heritability for diabetes microvascular (diabetic kidney disease and diabetic retinopathy) and macrovascular (cardiovascular events) complications. Heritability for diabetic kidney disease was estimated at 29%. The heritability estimate for microalbuminuria ranged from 24 to 60% and was 41% for macroalbuminuria. Heritability estimates of diabetic retinopathy ranged from 6 to 33%, depending on the phenotype definition. More severe diabetes retinopathy possessed higher genetic contributions. We show, for the first time, that rare variants account for much of the heritability of diabetic retinopathy. This study suggests that a large portion of the genetic risk of diabetes complications is yet to be discovered and emphasizes the need for additional genetic studies of diabetes complications.
Collapse
Affiliation(s)
- Juhyun Kim
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA
- Department of Biostatistics, University of Michigan, Ann Arbor, MI
| | - Aubrey Jensen
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA
| | - Seyoon Ko
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA
| | - Sridharan Raghavan
- University of Colorado School of Medicine, Aurora, CO
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| | - Lawrence S. Phillips
- Division of Endocrinology, Emory University School of Medicine, Atlanta, GA
- Atlanta Veterans Affairs Medical Center, Decatur, GA
| | - Adriana Hung
- Tennessee Valley Healthcare System and Vanderbilt University, Nashville, TN
| | - Yan Sun
- Department of Epidemiology, Emory University, Atlanta, GA
| | - Hua Zhou
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA
| | - Peter Reaven
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ
| | - Jin J. Zhou
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ
- Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
15
|
Type 1 Diabetes (T1D) and Latent Autoimmune Diabetes in Adults (LADA): The Difference Between a Honeymoon and a Holiday. Case Rep Endocrinol 2022; 2022:9363543. [PMID: 35355800 PMCID: PMC8960035 DOI: 10.1155/2022/9363543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 11/21/2022] Open
Abstract
Type 1 diabetes mellitus (T1D) is a chronic autoimmune disease in which destruction of the insulin-producing β-cells in the pancreatic islets requires regular lifelong insulin replacement therapy, the only lifesaving treatment available at this time. In young persons with a genetic predisposition, it usually manifests after being exposed to environmental triggers. A subtype of autoimmune diabetes mellitus (ADM) that typically occurs in adulthood is often referred to as latent autoimmune diabetes of adults (LADA). LADA is characterized by a milder process of β-cells destruction and less intensive insulin treatment, which may become necessary even many years after diagnosis. Genetic predisposition of T1D carries an increased risk for other autoimmune diseases, such as autoimmune thyroiditis, the most frequently associated condition, and pernicious anaemia (PA), present in approximately 4% of all individuals with T1D. Here, we describe the case of a 90-year-old woman with vitiligo and a mute medical history who was admitted to our University Hospital in Perugia with hyperglycaemia and severe anaemia due to vitamin B12 (VB12) depletion. A short time after setting the beginning treatment with a basal-bolus insulin regimen, her insulin requirement rapidly declined and treatment with sitagliptin, a dipeptidyl peptidase-4 inhibitor (DPP4), was started. A complete autoimmunity screening panel showed that GAD65 and intrinsic factor autoantibodies were positive.
Collapse
|
16
|
Suire CN, Hade MD. Extracellular Vesicles in Type 1 Diabetes: A Versatile Tool. Bioengineering (Basel) 2022; 9:105. [PMID: 35324794 PMCID: PMC8945706 DOI: 10.3390/bioengineering9030105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes is a chronic autoimmune disease affecting nearly 35 million people. This disease develops as T-cells continually attack the β-cells of the islets of Langerhans in the pancreas, which leads to β-cell death, and steadily decreasing secretion of insulin. Lowered levels of insulin minimize the uptake of glucose into cells, thus putting the body in a hyperglycemic state. Despite significant progress in the understanding of the pathophysiology of this disease, there is a need for novel developments in the diagnostics and management of type 1 diabetes. Extracellular vesicles (EVs) are lipid-bound nanoparticles that contain diverse content from their cell of origin and can be used as a biomarker for both the onset of diabetes and transplantation rejection. Furthermore, vesicles can be loaded with therapeutic cargo and delivered in conjunction with a transplant to increase cell survival and long-term outcomes. Crucially, several studies have linked EVs and their cargos to the progression of type 1 diabetes. As a result, gaining a better understanding of EVs would help researchers better comprehend the utility of EVs in regulating and understanding type 1 diabetes. EVs are a composition of biologically active components such as nucleic acids, proteins, metabolites, and lipids that can be transported to particular cells/tissues through the blood system. Through their varied content, EVs can serve as a flexible aid in the diagnosis and management of type 1 diabetes. In this review, we provide an overview of existing knowledge about EVs. We also cover the role of EVs in the pathogenesis, detection, and treatment of type 1 diabetes and the function of EVs in pancreas and islet β-cell transplantation.
Collapse
|
17
|
Corsenac P, Parent MÉ, Benedetti A, Richard H, Stäger S, Rousseau MC. Association between Bacillus Calmette-Guerin vaccination and type 1 diabetes in adolescence: A population-based birth cohort study in Quebec, Canada. Prev Med 2022; 154:106893. [PMID: 34798196 DOI: 10.1016/j.ypmed.2021.106893] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 10/12/2021] [Accepted: 11/14/2021] [Indexed: 10/19/2022]
Abstract
The Bacillus Calmette-Guerin (BCG) vaccine could reduce the incidence of type 1 diabetes through non-specific immunomodulation. Previous epidemiological studies, presenting some limitations, report no association. We examined this association of early life BCG vaccination and age at vaccination with type 1 diabetes incidence in adolescence in a large representative cohort in Quebec. The cohort included 387,704 individuals born in Quebec between 1970 and 1974 whose BCG vaccination status was determined from a provincial registry. Individuals were followed up from 1985 to their 19th birthday (maximum to 1993) for their use of physician services. Individuals were defined as type 1 diabetes cases if they had ≥4 related physician claims over a 2-year period, with at least 30 days between two claims. Cox proportional hazards regression was used to estimate the association of BCG vaccination and age at vaccination with type 1 diabetes. Covariates were selected based on a directed acyclic graph. Interaction according to sex was evaluated. A total of 178,133 (45.9%) individuals were vaccinated and 442 (0.11%) incident cases of type 1 diabetes were identified. The risk of type 1 diabetes was similar in vaccinated compared with unvaccinated individuals (adjusted hazard ratio = 1.06 [95% CI: 0.88-1.29]). There was no association with age at vaccination, and results did not differ by sex (Interaction, p = 0.60). Our results suggest that BCG vaccination does not prevent type 1 diabetes in adolescence.
Collapse
Affiliation(s)
- Philippe Corsenac
- Epidemiology and Biostatistics Unit, Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique (INRS), Laval, QC, Canada.
| | - Marie-Élise Parent
- Epidemiology and Biostatistics Unit, Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique (INRS), Laval, QC, Canada.
| | - Andrea Benedetti
- Respiratory Epidemiology and Clinical Research Unit, Research Institute of the McGill University Health Centre, McGill University, Montréal, QC, Canada; Department of Epidemiology, Biostatistics and Occupational Health, Faculty of Medicine, McGill University, Montréal, QC, Canada.
| | - Hugues Richard
- Epidemiology and Biostatistics Unit, Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique (INRS), Laval, QC, Canada.
| | - Simona Stäger
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique (INRS), Laval, QC, Canada.
| | - Marie-Claude Rousseau
- Epidemiology and Biostatistics Unit, Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique (INRS), Laval, QC, Canada.
| |
Collapse
|
18
|
Lima JEBF, Moreira NCS, Takahashi P, Xavier DJ, Sakamoto-Hojo ET. Oxidative Stress, DNA Damage, and Transcriptional Expression of DNA Repair and Stress Response Genes in Diabetes Mellitus. TRANSCRIPTOMICS IN HEALTH AND DISEASE 2022:341-365. [DOI: 10.1007/978-3-030-87821-4_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
19
|
Takahashi P, Xavier DJ, Lima JEBF, Evangelista AF, Collares CVA, Foss-Freitas MC, Rassi DM, Donadi EA, Passos GA, Sakamoto-Hojo ET. Transcript Expression Profiles and MicroRNA Regulation Indicate an Upregulation of Processes Linked to Oxidative Stress, DNA Repair, Cell Death, and Inflammation in Type 1 Diabetes Mellitus Patients. J Diabetes Res 2022; 2022:3511329. [PMID: 35155683 PMCID: PMC8825437 DOI: 10.1155/2022/3511329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/13/2022] [Indexed: 12/16/2022] Open
Abstract
Type 1 diabetes (T1D) arises from autoimmune-mediated destruction of insulin-producing β-cells leading to impaired insulin secretion and hyperglycemia. T1D is accompanied by DNA damage, oxidative stress, and inflammation, although there is still scarce information about the oxidative stress response and DNA repair in T1D pathogenesis. We used the microarray method to assess mRNA expression profiles in peripheral blood mononuclear cells (PBMCs) of 19 T1D patients compared to 11 controls and identify mRNA targets of microRNAs that were previously reported for T1D patients. We found 277 differentially expressed genes (220 upregulated and 57 downregulated) in T1D patients compared to controls. Analysis by gene sets (GSA and GSEA) showed an upregulation of processes linked to ROS generation, oxidative stress, inflammation, cell death, ER stress, and DNA repair in T1D patients. Besides, genes related to oxidative stress responses and DNA repair (PTGS2, ATF3, FOSB, DUSP1, and TNFAIP3) were found to be targets of four microRNAs (hsa-miR-101, hsa-miR148a, hsa-miR-27b, and hsa-miR-424). The expression levels of these mRNAs and microRNAs were confirmed by qRT-PCR. Therefore, the present study on differential expression profiles indicates relevant biological functions related to oxidative stress response, DNA repair, inflammation, and apoptosis in PBMCs of T1D patients relative to controls. We also report new insights regarding microRNA-mRNA interactions, which may play important roles in the T1D pathogenesis.
Collapse
Affiliation(s)
- Paula Takahashi
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, 14049900, SP, Brazil
| | - Danilo J. Xavier
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, 14049900, SP, Brazil
| | - Jessica E. B. F. Lima
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, 14049900, SP, Brazil
| | | | - Cristhianna V. A. Collares
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, 14049900, SP, Brazil
- Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Maria C. Foss-Freitas
- Division of Endocrinology, Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Diane M. Rassi
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Eduardo A. Donadi
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, 14049900, SP, Brazil
- Division of Clinical Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Geraldo A. Passos
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, 14049900, SP, Brazil
- Laboratory of Genetics and Molecular Biology, Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Elza T. Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, 14049900, SP, Brazil
- Department of Biology, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| |
Collapse
|
20
|
Pellenz FM, Dieter C, Duarte GCK, Canani LH, de Souza BM, Crispim D. The rs2304256 Polymorphism in TYK2 Gene Is Associated with Protection for Type 1 Diabetes Mellitus. Diabetes Metab J 2021; 45:899-908. [PMID: 34225445 PMCID: PMC8640150 DOI: 10.4093/dmj.2020.0194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/04/2020] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Tyrosine kinase 2 (TYK2) is a candidate gene for type 1 diabetes mellitus (T1DM) since it plays an important role in regulating apoptotic and pro-inflammatory pathways in pancreatic β-cells through modulation of the type I interferon signaling pathway. The rs2304256 single nucleotide polymorphism (SNP) in TYK2 gene has been associated with protection for different autoimmune diseases. However, to date, only two studies have evaluated the association between this SNP and T1DM, with discordant results. This study thus aimed to investigate the association between the TYK2 rs2304256 SNP and T1DM in a Southern Brazilian population. METHODS This case-control study comprised 478 patients with T1DM and 518 non-diabetic subjects. The rs2304256 (C/A) SNP was genotyped by real-time polymerase chain reaction technique using TaqMan minor groove binder (MGB) probes. RESULTS Genotype and allele frequencies of the rs2304256 SNP differed between T1DM patients and non-diabetic subjects (P<0.0001 and P=0.001, respectively). Furthermore, the A allele was associated with protection against T1DM under recessive (odds ratio [OR], 0.482; 95% confidence interval [CI], 0.288 to 0.806) and additive (OR, 0.470; 95% CI, 0.278 to 0.794) inheritance models, adjusting for human leukocyte antigen (HLA) DR/DQ genotypes, gender, and ethnicity. CONCLUSION The A/A genotype of TYK2 rs2304256 SNP is associated with protection against T1DM in a Southern Brazilian population.
Collapse
Affiliation(s)
- Felipe Mateus Pellenz
- Endocrinology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Federal University of Rio Grande do Sul, Faculty of Medicine, Porto Alegre, Brazil
| | - Cristine Dieter
- Endocrinology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Federal University of Rio Grande do Sul, Faculty of Medicine, Porto Alegre, Brazil
| | - Guilherme Coutinho Kullmann Duarte
- Endocrinology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Federal University of Rio Grande do Sul, Faculty of Medicine, Porto Alegre, Brazil
| | - Luís Henrique Canani
- Endocrinology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Federal University of Rio Grande do Sul, Faculty of Medicine, Porto Alegre, Brazil
| | - Bianca Marmontel de Souza
- Endocrinology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Federal University of Rio Grande do Sul, Faculty of Medicine, Porto Alegre, Brazil
| | - Daisy Crispim
- Endocrinology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Federal University of Rio Grande do Sul, Faculty of Medicine, Porto Alegre, Brazil
- Corresponding author: Daisy Crispim https://orcid.org/0000-0001-5095-9269 Endocrinology Division, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos 2350, prédio 12, 4° Andar, Porto Alegre 90035-003, Brazil E-mail:
| |
Collapse
|
21
|
Chen J, Zhai C, Wang Z, Li R, Wu W, Hou K, Alzogool M, Wang Y, Cong H. The susceptibility of SERPINE1 rs1799889 SNP in diabetic vascular complications: a meta-analysis of fifty-one case-control studies. BMC Endocr Disord 2021; 21:195. [PMID: 34592988 PMCID: PMC8482645 DOI: 10.1186/s12902-021-00837-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 08/10/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The serine protease inhibitor-1 (SERPINE1) rs1799889 single nucleotide polymorphism (SNP) has been constantly associated with diabetes mellitus (DM) and its vascular complications. The aim of this meta-analysis was to evaluate this association with combined evidences. METHODS The systematic search was performed for studies published up to March 2021 which assess the associations between SERPINE1 rs1799889 SNP and the risks of DM, diabetic retinopathy (DR), diabetic cardiovascular disease (CVD) and diabetic nephropathy (DN). Only case-control studies were identified, and the linkage between SERPINE1 rs1799889 polymorphism and diabetic vascular risks were evaluated using genetic models. RESULTS 51 comparisons were enrolled. The results revealed a significant association with diabetes risk in overall population (allelic: OR = 1.34, 95 % CI = 1.14-1.57, homozygous: OR = 1.66, 95 % CI = 1.23-2.14, heterozygous: OR = 1.35, 95 % CI = 1.08-1.69, dominant: OR = 1.49, 95 % CI = 1.18-1.88, recessive: OR = 1.30, 95 % CI = 1.06-1.59) as well as in Asian descents (allelic: OR = 1.45, 95 % CI = 1.16-1.82, homozygous: OR = 1.88, 95 % CI = 1.29-2.75, heterozygous: OR = 1.47, 95 % CI = 1.08-2.00, dominant: OR = 1.64, 95 % CI = 1.21-2.24, recessive: OR = 1.46, 95 % CI = 1.09-1.96). A significant association was observed with DR risk (homozygous: OR = 1.25, 95 % CI = 1.01-1.56, recessive: OR = 1.20, 95 % CI = 1.01-1.43) for overall population, as for the European subgroup (homozygous: OR = 1.32, 95 % CI = 1.02-1.72, recessive: OR = 1.38, 95 % CI = 1.11-1.71). A significant association were shown with DN risk for overall population (allelic: OR = 1.48, 95 % CI = 1.15-1.90, homozygous: OR = 1.92, 95 % CI = 1.26-2.95, dominant: OR = 1.41, 95 % CI = 1.01-1.97, recessive: OR = 1.78, 95 % CI = 1.27-2.51) and for Asian subgroup (allelic: OR = 1.70, 95 % CI = 1.17-2.47, homozygous: OR = 2.46, 95 % CI = 1.30-4.66, recessive: OR = 2.24, 95 % CI = 1.40-3.59) after ethnicity stratification. No obvious association was implied with overall diabetic CVD risk in any genetic models, or after ethnicity stratification. CONCLUSIONS SERPINE1 rs1799889 4G polymorphism may outstand for serving as a genetic synergistic factor in overall DM and DN populations, positively for individuals with Asian descent. The association of SERPINE1 rs1799889 SNP and DR or diabetic CVD risks was not revealed.
Collapse
Affiliation(s)
- JingYi Chen
- School of Medicine, NanKai University, Weijin Road No. 94, Nankai District, 300071 Tianjin, China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Gansu Road No. 4, Heping District, 300020 Tianjin, China
| | - ChuanNan Zhai
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang south Road No. 291, Jinnan District, 300350 Tianjin, China
| | - ZhiQian Wang
- Department of Optometry, Shenyang Eye Institute, The 4th People’s Hospital of Shenyang, No 20. Huanghe South Avenue, Huanggu District, 110031 Shenyang, Liaoning China
| | - Rui Li
- Tianjin GongAn Hospital, Nanjing Road No. 78, Heping District, 300042 Tianjin, China
| | - WenJing Wu
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Gansu Road No. 4, Heping District, 300020 Tianjin, China
| | - Kai Hou
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang south Road No. 291, Jinnan District, 300350 Tianjin, China
| | - Mohammad Alzogool
- School of Medicine, NanKai University, Weijin Road No. 94, Nankai District, 300071 Tianjin, China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Gansu Road No. 4, Heping District, 300020 Tianjin, China
| | - Yan Wang
- School of Medicine, NanKai University, Weijin Road No. 94, Nankai District, 300071 Tianjin, China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Gansu Road No. 4, Heping District, 300020 Tianjin, China
| | - HongLiang Cong
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang south Road No. 291, Jinnan District, 300350 Tianjin, China
| |
Collapse
|
22
|
Polymorphisms in GLIS3 and susceptibility to diabetes mellitus: A systematic review and meta-analysis. Meta Gene 2021. [DOI: 10.1016/j.mgene.2021.100898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
23
|
Bauer W, Gyenesei A, Krętowski A. The Multifactorial Progression from the Islet Autoimmunity to Type 1 Diabetes in Children. Int J Mol Sci 2021; 22:7493. [PMID: 34299114 PMCID: PMC8305179 DOI: 10.3390/ijms22147493] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/04/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Type 1 Diabetes (T1D) results from autoimmune destruction of insulin producing pancreatic ß-cells. This disease, with a peak incidence in childhood, causes the lifelong need for insulin injections and necessitates careful monitoring of blood glucose levels. However, despite the current insulin therapies, it still shortens life expectancy due to complications affecting multiple organs. Recently, the incidence of T1D in childhood has increased by 3-5% per year in most developed Western countries. The heterogeneity of the disease process is supported by the findings of follow-up studies started early in infancy. The development of T1D is usually preceded by the appearance of autoantibodies targeted against antigens expressed in the pancreatic islets. The risk of T1D increases significantly with an increasing number of positive autoantibodies. The order of autoantibody appearance affects the disease risk. Genetic susceptibility, mainly defined by the human leukocyte antigen (HLA) class II gene region and environmental factors, is important in the development of islet autoimmunity and T1D. Environmental factors, mainly those linked to the changes in the gut microbiome as well as several pathogens, especially viruses, and diet are key modulators of T1D. The aim of this paper is to expand the understanding of the aetiology and pathogenesis of T1D in childhood by detailed description and comparison of factors affecting the progression from the islet autoimmunity to T1D in children.
Collapse
Affiliation(s)
- Witold Bauer
- Clinical Research Centre, Medical University of Białystok, Marii Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (A.G.); (A.K.)
| | - Attila Gyenesei
- Clinical Research Centre, Medical University of Białystok, Marii Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (A.G.); (A.K.)
- Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, 7624 Pécs, Hungary
| | - Adam Krętowski
- Clinical Research Centre, Medical University of Białystok, Marii Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (A.G.); (A.K.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Białystok, Marii Skłodowskiej-Curie 24a, 15-276 Białystok, Poland
| |
Collapse
|
24
|
Vojtková J, Kolková Z, Motyková K, Kostková M, Suroviaková S, Grendár M, Bánovčin P. An association between fibrinogen gene polymorphisms and diabetic peripheral neuropathy in young patients with type 1 diabetes. Mol Biol Rep 2021; 48:4397-4404. [PMID: 34061326 DOI: 10.1007/s11033-021-06455-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/27/2021] [Indexed: 11/30/2022]
Abstract
In complex etiopathogenesis of diabetic peripheral neuropathy (DPN), hemostatic dysfunction and subclinical inflammation play a possible role. Fibrinogen is involved in both the hemostatic and inflammatory pathways, so we hypothesize that fibrinogen gene polymorphisms might be associated with DPN. A total of 127 young patients with type 1 diabetes (T1D) (average age, 18.5 ± 4.65 years; average diabetes duration, 14.5 ± 2.26 years) and 90 healthy controls were enrolled into the study. Basic biochemical and coagulation parameters were measured and gene polymorphisms of fibrinogen alpha (rs6050) and beta (rs1800790) were established. DPN was diagnosed in 38 diabetic patients by neurological examination. AA genotype and A allele of rs1800790 polymorphism of fibrinogen beta were associated with increased risk of DPN (odds ratio [OR] 4.537, 95% confidence interval [95CI] 1.14-19.94, p = 0.019 and OR 1.958, 95CI 1.038-3.675, p = 0.029, respectively). No association was found between DPN and rs6050 gene polymorphisms. Plasma fibrinogen concentration significantly correlated with HbA1c (Spearman's correlation coefficient [r] = 0.54) and HDL cholesterol (r = - 0.67). A allele and AA genotype of rs1800790 seem to be associated with DPN in young patients with T1D. Further studies are appropriate to elucidate the role of fibrinogen gene polymorphisms in the complex etiology of DPN.
Collapse
Affiliation(s)
- Jarmila Vojtková
- Department of Pediatrics, Jessenius Faculty of Medicine and University Hospital, Comenius University in Bratislava, Kollárova 2, 036 01, Martin, Slovakia
| | - Zuzana Kolková
- Department of Pediatrics, Jessenius Faculty of Medicine and University Hospital, Comenius University in Bratislava, Kollárova 2, 036 01, Martin, Slovakia.,Jessenius Faculty of Medicine, Biomedical Center Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Katarína Motyková
- Department of Pediatrics, Jessenius Faculty of Medicine and University Hospital, Comenius University in Bratislava, Kollárova 2, 036 01, Martin, Slovakia
| | - Martina Kostková
- Department of Pediatrics, Jessenius Faculty of Medicine and University Hospital, Comenius University in Bratislava, Kollárova 2, 036 01, Martin, Slovakia
| | - Stanislava Suroviaková
- Department of Pediatrics, Jessenius Faculty of Medicine and University Hospital, Comenius University in Bratislava, Kollárova 2, 036 01, Martin, Slovakia.
| | - Marián Grendár
- Jessenius Faculty of Medicine, Bioinformatic Center, Biomedical Center Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Peter Bánovčin
- Department of Pediatrics, Jessenius Faculty of Medicine and University Hospital, Comenius University in Bratislava, Kollárova 2, 036 01, Martin, Slovakia
| |
Collapse
|
25
|
Illing PT, van Hateren A, Darley R, Croft NP, Mifsud NA, King S, Kostenko L, Bharadwaj M, McCluskey J, Elliott T, Purcell AW. Kinetics of Abacavir-Induced Remodelling of the Major Histocompatibility Complex Class I Peptide Repertoire. Front Immunol 2021; 12:672737. [PMID: 34093574 PMCID: PMC8170132 DOI: 10.3389/fimmu.2021.672737] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/26/2021] [Indexed: 12/14/2022] Open
Abstract
Abacavir hypersensitivity syndrome can occur in individuals expressing the HLA-B*57:01 major histocompatibility complex class I allotype when utilising the drug abacavir as a part of their anti-retroviral regimen. The drug is known to bind within the HLA-B*57:01 antigen binding cleft, leading to the selection of novel self-peptide ligands, thus provoking life-threatening immune responses. However, the sub-cellular location of abacavir binding and the mechanics of altered peptide selection are not well understood. Here, we probed the impact of abacavir on the assembly of HLA-B*57:01 peptide complexes. We show that whilst abacavir had minimal impact on the maturation or average stability of HLA-B*57:01 molecules, abacavir was able to differentially enhance the formation, selectively decrease the dissociation, and alter tapasin loading dependency of certain HLA-B*57:01-peptide complexes. Our data reveals a spectrum of abacavir mediated effects on the immunopeptidome which reconciles the heterogeneous functional T cell data reported in the literature.
Collapse
Affiliation(s)
- Patricia T. Illing
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Andy van Hateren
- Institute for Life Sciences and Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Rachel Darley
- Institute for Life Sciences and Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Nathan P. Croft
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Nicole A. Mifsud
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Samuel King
- Institute for Life Sciences and Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Lyudmila Kostenko
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Mandvi Bharadwaj
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - James McCluskey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Tim Elliott
- Institute for Life Sciences and Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Anthony W. Purcell
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
26
|
Shi Q, Yao H. Signature RNAS and related regulatory roles in type 1 diabetes mellitus based on competing endogenous RNA regulatory network analysis. BMC Med Genomics 2021; 14:133. [PMID: 34006268 PMCID: PMC8130321 DOI: 10.1186/s12920-021-00931-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 03/04/2021] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Our study aimed to investigate signature RNAs and their potential roles in type 1 diabetes mellitus (T1DM) using a competing endogenous RNA regulatory network analysis. METHODS Expression profiles of GSE55100, deposited from peripheral blood mononuclear cells of 12 T1DM patients and 10 normal controls, were downloaded from the Gene Expression Omnibus to uncover differentially expressed long non-coding RNAs (lncRNAs), mRNAs, and microRNAs (miRNAs). The ceRNA regulatory network was constructed, then functional and pathway enrichment analysis was conducted. AT1DM-related ceRNA regulatory network was established based on the Human microRNA Disease Database to carry out pathway enrichment analysis. Meanwhile, the T1DM-related pathways were retrieved from the Comparative Toxicogenomics Database (CTD). RESULTS In total, 847 mRNAs, 41 lncRNAs, and 38 miRNAs were significantly differentially expressed. The ceRNA regulatory network consisted of 12 lncRNAs, 10 miRNAs, and 24 mRNAs. Two miRNAs (hsa-miR-181a and hsa-miR-1275) were screened as T1DM-related miRNAs to build the T1DM-related ceRNA regulatory network, in which genes were considerably enriched in seven pathways. Moreover, three overlapping pathways, including the phosphatidylinositol signaling system (involving PIP4K2A, INPP4A, PIP4K2C, and CALM1); dopaminergic synapse (involving CALM1 and PPP2R5C); and the insulin signaling pathway (involving CBLB and CALM1) were revealed by comparing with T1DM-related pathways in the CTD, which involved four lncRNAs (LINC01278, TRG-AS1, MIAT, and GAS5-AS1). CONCLUSION The identified signature RNAs may serve as important regulators in the pathogenesis of T1DM.
Collapse
Affiliation(s)
- Qinghong Shi
- Department of Clinical Laboratory, The Third Hospital of Jilin University, No. 126, Xiantai Street, Changchun, 130033 Jilin China
| | - Hanxin Yao
- Department of Clinical Laboratory, The First Hospital of Jilin University, No. 1, Xinmin Street, Chaoyang District, Changchun, 130021 Jilin China
| |
Collapse
|
27
|
Shapiro MR, Thirawatananond P, Peters L, Sharp RC, Ogundare S, Posgai AL, Perry DJ, Brusko TM. De-coding genetic risk variants in type 1 diabetes. Immunol Cell Biol 2021; 99:496-508. [PMID: 33483996 PMCID: PMC8119379 DOI: 10.1111/imcb.12438] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/08/2021] [Accepted: 01/20/2021] [Indexed: 12/13/2022]
Abstract
The conceptual basis for a genetic predisposition underlying the risk for developing type 1 diabetes (T1D) predates modern human molecular genetics. Over half of the genetic risk has been attributed to the human leukocyte antigen (HLA) class II gene region and to the insulin (INS) gene locus - both thought to confer direction of autoreactivity and tissue specificity. Notwithstanding, questions still remain regarding the functional contributions of a vast array of minor polygenic risk variants scattered throughout the genome that likely influence disease heterogeneity and clinical outcomes. Herein, we summarize the available literature related to the T1D-associated coding variants defined at the time of this review, for the genes PTPN22, IFIH1, SH2B3, CD226, TYK2, FUT2, SIRPG, CTLA4, CTSH and UBASH3A. Data from genotype-selected human cohorts are summarized, and studies from the non-obese diabetic (NOD) mouse are presented to describe the functional impact of these variants in relation to innate and adaptive immunity as well as to β-cell fragility, with expression profiles in tissues and peripheral blood highlighted. The contribution of each variant to progression through T1D staging, including environmental interactions, are discussed with consideration of how their respective protein products may serve as attractive targets for precision medicine-based therapeutics to prevent or suspend the development of T1D.
Collapse
Affiliation(s)
- Melanie R Shapiro
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Puchong Thirawatananond
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Leeana Peters
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Robert C Sharp
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Similoluwa Ogundare
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Daniel J Perry
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Todd M Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
- Department of Pediatrics, College of Medicine, Diabetes Institute, University of Florida, Gainesville, FL, 32610, USA
| |
Collapse
|
28
|
Alcazar O, Hernandez LF, Nakayasu ES, Nicora CD, Ansong C, Muehlbauer MJ, Bain JR, Myer CJ, Bhattacharya SK, Buchwald P, Abdulreda MH. Parallel Multi-Omics in High-Risk Subjects for the Identification of Integrated Biomarker Signatures of Type 1 Diabetes. Biomolecules 2021; 11:383. [PMID: 33806609 PMCID: PMC7999903 DOI: 10.3390/biom11030383] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Biomarkers are crucial for detecting early type-1 diabetes (T1D) and preventing significant β-cell loss before the onset of clinical symptoms. Here, we present proof-of-concept studies to demonstrate the potential for identifying integrated biomarker signature(s) of T1D using parallel multi-omics. METHODS Blood from human subjects at high risk for T1D (and healthy controls; n = 4 + 4) was subjected to parallel unlabeled proteomics, metabolomics, lipidomics, and transcriptomics. The integrated dataset was analyzed using Ingenuity Pathway Analysis (IPA) software for disturbances in the at-risk subjects compared to controls. RESULTS The final quadra-omics dataset contained 2292 proteins, 328 miRNAs, 75 metabolites, and 41 lipids that were detected in all samples without exception. Disease/function enrichment analyses consistently indicated increased activation, proliferation, and migration of CD4 T-lymphocytes and macrophages. Integrated molecular network predictions highlighted central involvement and activation of NF-κB, TGF-β, VEGF, arachidonic acid, and arginase, and inhibition of miRNA Let-7a-5p. IPA-predicted candidate biomarkers were used to construct a putative integrated signature containing several miRNAs and metabolite/lipid features in the at-risk subjects. CONCLUSIONS Preliminary parallel quadra-omics provided a comprehensive picture of disturbances in high-risk T1D subjects and highlighted the potential for identifying associated integrated biomarker signatures. With further development and validation in larger cohorts, parallel multi-omics could ultimately facilitate the classification of T1D progressors from non-progressors.
Collapse
Affiliation(s)
- Oscar Alcazar
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (O.A.); (L.F.H.)
| | - Luis F. Hernandez
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (O.A.); (L.F.H.)
| | - Ernesto S. Nakayasu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (E.S.N.); (C.D.N.); (C.A.)
| | - Carrie D. Nicora
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (E.S.N.); (C.D.N.); (C.A.)
| | - Charles Ansong
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA; (E.S.N.); (C.D.N.); (C.A.)
| | - Michael J. Muehlbauer
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA; (M.J.M.); (J.R.B.)
| | - James R. Bain
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27701, USA; (M.J.M.); (J.R.B.)
| | - Ciara J. Myer
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.J.M.); (S.K.B.)
- Miami Integrative Metabolomics Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sanjoy K. Bhattacharya
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.J.M.); (S.K.B.)
- Miami Integrative Metabolomics Research Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Peter Buchwald
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (O.A.); (L.F.H.)
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Midhat H. Abdulreda
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (O.A.); (L.F.H.)
- Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.J.M.); (S.K.B.)
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
29
|
Sanz CR, Sevane N, Pérez-Alenza MD, Valero-Lorenzo M, Dunner S. Polymorphisms in canine immunoglobulin heavy chain gene cluster: a double-edged sword for diabetes mellitus in the dog. Anim Genet 2021; 52:333-341. [PMID: 33621396 DOI: 10.1111/age.13047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
Insulin deficiency diabetes (IDD) in dogs is an endocrine disease similar to human type 1 diabetes. There are breeds more commonly affected, such as Yorkshire Terrier and Samoyed, suggesting an underlying genetic component. However, the genetic basis for canine diabetes mellitus (DM) is not fully established. We conducted both whole-genome scans for selection signatures and GWASs to compare the genomes of 136 dogs belonging to 29 breeds previously described at low or high risk for developing DM. Candidate variants were tested in dogs with a diagnosis of IDD and controls attending the Complutense Veterinary Teaching Hospital. The only genomic region under selection (CFA8:72 700 000-74 600 000; CanFam3.1) retrieved by our analyses is included in the immunoglobulin heavy chain gene cluster, which has already been related to human human type 1 diabetes susceptibility. This region contains two non-synonymous variants, rs852072969 and rs851728071, showing significant associations with high or low risk for IDD, respectively. The first variant, rs852072969, alters a protein poorly characterised in the dog. In contrast, rs851728071 was predicted to block the synthesis of an immunoglobulin variable (V) domain in breeds at low risk for DM. Although a large and diverse V gene repertoire is thought to offer a fitness advantage, we suggest that rs851728071 prevents the formation of an auto-reactive immunoglobulin V domain probably involved in the pathophysiology of IDD and, thus, decreases the risk for the disease. These results should be interpreted with caution until the functional roles of the proposed variants have been proved in larger studies.
Collapse
Affiliation(s)
- C R Sanz
- Department of Animal Production, Veterinary Faculty, Complutense University of Madrid, Madrid, 28040, Spain
| | - N Sevane
- Department of Animal Production, Veterinary Faculty, Complutense University of Madrid, Madrid, 28040, Spain
| | - M D Pérez-Alenza
- Department of Animal Medicine, Surgery and Pathology, Veterinary Faculty, Complutense University of Madrid, Madrid, 28040, Spain
| | - M Valero-Lorenzo
- Department of Animal Production, Veterinary Faculty, Complutense University of Madrid, Madrid, 28040, Spain
| | - S Dunner
- Department of Animal Production, Veterinary Faculty, Complutense University of Madrid, Madrid, 28040, Spain
| |
Collapse
|
30
|
Battaglia M, Buckner JH, Levings MK, Richardson SJ, Wong FS, Tree TI. Identifying the 'Achilles heel' of type 1 diabetes. Clin Exp Immunol 2021; 204:167-178. [PMID: 33368173 DOI: 10.1111/cei.13570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 12/22/2022] Open
Abstract
When Thetis dipped her son Achilles into the River Styx to make him immortal, she held him by the heel, which was not submerged, and thus created a weak spot that proved deadly for Achilles. Millennia later, Achilles heel is part of today's lexicon meaning an area of weakness or a vulnerable spot that causes failure. Also implied is that an Achilles heel is often missed, forgotten or under-appreciated until it is under attack, and then failure is fatal. Paris killed Achilles with an arrow 'guided by the Gods'. Understanding the pathogenesis of type 1 diabetes (T1D) in order to direct therapy for prevention and treatment is a major goal of research into T1D. At the International Congress of the Immunology of Diabetes Society, 2018, five leading experts were asked to present the case for a particular cell/element that could represent 'the Achilles heel of T1D'. These included neutrophils, B cells, CD8+ T cells, regulatory CD4+ T cells, and enteroviruses, all of which have been proposed to play an important role in the pathogenesis of type 1 diabetes. Did a single entity emerge as 'the' Achilles heel of T1D? The arguments are summarized here, to make this case.
Collapse
Affiliation(s)
- M Battaglia
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - J H Buckner
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - M K Levings
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - S J Richardson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - F S Wong
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - T I Tree
- Department of Immunobiology, School of Immunology and Microbial Sciences (SIMS), King's College London, London, UK.,NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
31
|
Type 1 diabetes: genes associated with disease development. Cent Eur J Immunol 2021; 45:439-453. [PMID: 33658892 PMCID: PMC7882399 DOI: 10.5114/ceji.2020.103386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 01/02/2020] [Indexed: 11/17/2022] Open
Abstract
Type 1 diabetes (T1D) is the third most common autoimmune disease which develops due to genetic and environmental risk factors. Based on the World Health Organization (WHO) report from 2014 the number of people suffering from all types of diabetes ascended to 422 million, compared to 108 million in 1980. It was calculated that this number will double by the end of 2030. In 2015 American Diabetes Association (ADA) announced that 30.3 million Americans (that is 9.4% of the overall population) had diabetes of which only approximately 1.25 million had T1D. Nowadays, T1D represents roughly 10% of adult diabetes cases total. Multiple genetic abnormalities at different loci have been found to contribute to type 1 diabetes development. The analysis of genome-wide association studies (GWAS) of T1D has identified over 50 susceptible regions (and genes within these regions). Many of these regions are defined by single nucleotide polymorphisms (SNPs) but molecular mechanisms through which they increase or lower the risk of diabetes remain unknown. Genetic factors (in existence since birth) can be detected long before the emergence of immunological or clinical markers. Therefore, a comprehensive understanding of the multiple genetic factors underlying T1D is extremely important for further clinical trials and development of personalized medicine for diabetic patients. We present an overview of current studies and information about regions in the human genome associated with T1D. Moreover, we also put forward information about epigenetic modifications, non-coding RNAs and environmental factors involved in T1D development and onset.
Collapse
|
32
|
Genetics of canine diabetes mellitus part 1: Phenotypes of disease. Vet J 2021; 270:105611. [PMID: 33641807 DOI: 10.1016/j.tvjl.2021.105611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 01/09/2023]
Abstract
This two-part article discusses the mechanisms by which genetic variation can influence the risk of complex diseases, with a focus on canine diabetes mellitus. In Part 1, presented here, the importance of accurate methods for classifying different types of diabetes will be discussed, since this underpins the selection of cases and controls for genetic studies. Part 2 will focus on our current understanding of the genes involved in diabetes risk, and the way in which new genome sequencing technologies are poised to reveal new diabetes genes in veterinary species.
Collapse
|
33
|
Denyer AL, Catchpole B, Davison LJ. Genetics of canine diabetes mellitus part 2: Current understanding and future directions. Vet J 2021; 270:105612. [PMID: 33641811 DOI: 10.1016/j.tvjl.2021.105612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 02/08/2023]
Abstract
Part 1 of this 2-part review outlined the importance of disease classification in diabetes genetic studies, as well as the ways in which genetic variants may contribute to risk of a complex disease within an individual, or within a particular group of individuals. Part 2, presented here, describes in more detail our current understanding of the genetics of canine diabetes mellitus compared to our knowledge of the human disease. Ongoing work to improve our knowledge, using new technologies, is also introduced.
Collapse
Affiliation(s)
- Alice L Denyer
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hatfield, UK
| | - Brian Catchpole
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hatfield, UK
| | - Lucy J Davison
- Department of Clinical Sciences and Services, Royal Veterinary College, Hatfield, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| | | |
Collapse
|
34
|
Calabrese CM, Valentini A, Calabrese G. Gut Microbiota and Type 1 Diabetes Mellitus: The Effect of Mediterranean Diet. Front Nutr 2021; 7:612773. [PMID: 33521039 PMCID: PMC7838384 DOI: 10.3389/fnut.2020.612773] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic autoimmune disease resulting from a complex interplay between genetic susceptibility and environmental factors. Regarding the latter, gut microbiota has a pivotal role in the pathogenesis of T1DM, by affecting intestinal permeability, molecular mimicry, and modulating innate and adaptive immune system, as described in several previous studies. The composition of the gut microbiota is largely influenced by diet. Some observational studies have shown that a low fiber intake is associated with the development of many inflammatory and immune-mediated diseases. In this context, the Mediterranean diet (MD), which is based on high consumption of cereals (preferably as whole grains), legumes, nuts, vegetables, fruits, olive oil, and fish, could play a protective role. Many of the characteristic components of MD have functional characteristics with positive effects on health and well-being. Eating habits are the main significant determinants of the microbial multiplicity of the intestine and the food components influence both microbial populations and their metabolic activities from the early stages of life. Moreover, food metabolites influence the immune response. The intestine is considered the primary site where food metabolites mediate their effects, through epithelial integrity or mucosal immunity. The compromised epithelial integrity allows the translocation of bacteria and/or the diffusion of their products, such as food antigens and lipopolysaccharides, from the intestinal lumen to the tissues, which could enhance the stimulation of immune cells, contributing to the pathogenesis of autoimmune diseases, such as T1DM. The intake of a high amount of fiber and therefore of prebiotics with MD allows the microbiota to have a good microbial balance. Moreover, as more dietary fibers are ingested, a higher amount of short-chain fatty acids (SCFAs) is produced by anaerobic gut microbiota, promoting gut homeostasis, to which also contribute tryptophan metabolites and omega-3-fatty acids. Furthermore, the higher intake of polyunsaturated fatty acids and omega-3-fatty-acids contribute to a better metabolic control. In this review we report the relationship between gut microbiota and T1DM and we explore the effects of Mediterranean diet on microbiota as a potential therapeutic strategy, aimed at preventing or delaying progression of T1DM and its complications.
Collapse
Affiliation(s)
| | - Alessia Valentini
- Dipartimento di Medicina Interna, Ospedale Madre Giuseppina Vannini, Rome, Italy
| | - Giorgio Calabrese
- Dipartimento di Scienze e Innovazione Tecnologica, Università del Piemonte Orientale, Alessandria, Italy
| |
Collapse
|
35
|
Ramos-Rodríguez M, Pérez-González B, Pasquali L. The β-Cell Genomic Landscape in T1D: Implications for Disease Pathogenesis. Curr Diab Rep 2021; 21:1. [PMID: 33387073 PMCID: PMC7778620 DOI: 10.1007/s11892-020-01370-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/19/2020] [Indexed: 11/15/2022]
Abstract
PURPOSE OF REVIEW Type 1 diabetes (T1D) develops as a consequence of a combination of genetic predisposition and environmental factors. Combined, these events trigger an autoimmune disease that results in progressive loss of pancreatic β cells, leading to insulin deficiency. This article reviews the current knowledge on the genetics of T1D with a specific focus on genetic variation in pancreatic islet regulatory networks and its implication to T1D risk and disease development. RECENT FINDINGS Accumulating evidence suggest an active role of β cells in T1D pathogenesis. Based on such observation several studies aimed in mapping T1D risk variants acting at the β cell level. Such studies unravel T1D risk loci shared with type 2 diabetes (T2D) and T1D risk variants potentially interfering with β-cell responses to external stimuli. The characterization of regulatory genomics maps of disease-relevant states and cell types can be used to elucidate the mechanistic role of β cells in the pathogenesis of T1D.
Collapse
Affiliation(s)
- Mireia Ramos-Rodríguez
- Endocrine Regulatory Genomics, Department of Experimental & Health Sciences, University Pompeu Fabra, 08003, Barcelona, Spain
| | - Beatriz Pérez-González
- Endocrine Regulatory Genomics, Department of Experimental & Health Sciences, University Pompeu Fabra, 08003, Barcelona, Spain
| | - Lorenzo Pasquali
- Endocrine Regulatory Genomics, Department of Experimental & Health Sciences, University Pompeu Fabra, 08003, Barcelona, Spain.
| |
Collapse
|
36
|
Sayed S, Nabi AHMN. Diabetes and Genetics: A Relationship Between Genetic Risk Alleles, Clinical Phenotypes and Therapeutic Approaches. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1307:457-498. [PMID: 32314317 DOI: 10.1007/5584_2020_518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Unveiling human genome through successful completion of Human Genome Project and International HapMap Projects with the advent of state of art technologies has shed light on diseases associated genetic determinants. Identification of mutational landscapes such as copy number variation, single nucleotide polymorphisms or variants in different genes and loci have revealed not only genetic risk factors responsible for diseases but also region(s) playing protective roles. Diabetes is a global health concern with two major types - type 1 diabetes (T1D) and type 2 diabetes (T2D). Great progress in understanding the underlying genetic predisposition to T1D and T2D have been made by candidate gene studies, genetic linkage studies, genome wide association studies with substantial number of samples. Genetic information has importance in predicting clinical outcomes. In this review, we focus on recent advancement regarding candidate gene(s) associated with these two traits along with their clinical parameters as well as therapeutic approaches perceived. Understanding genetic architecture of these disease traits relating clinical phenotypes would certainly facilitate population stratification in diagnosing and treating T1D/T2D considering the doses and toxicity of specific drugs.
Collapse
Affiliation(s)
- Shomoita Sayed
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh
| | - A H M Nurun Nabi
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, Bangladesh.
| |
Collapse
|
37
|
Dieter C, Lemos NE, Corrêa NRDF, Assmann TS, Crispim D. The Impact of lncRNAs in Diabetes Mellitus: A Systematic Review and In Silico Analyses. Front Endocrinol (Lausanne) 2021; 12:602597. [PMID: 33815273 PMCID: PMC8018579 DOI: 10.3389/fendo.2021.602597] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 02/15/2021] [Indexed: 12/17/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are non-coding transcripts that have emerged as one of the largest and diverse RNA families that regulate gene expression. Accumulating evidence has suggested a number of lncRNAs are involved in diabetes mellitus (DM) pathogenesis. However, results about lncRNA expressions in DM patients are still inconclusive. Thus, we performed a systematic review of the literature on the subject followed by bioinformatics analyses to better understand which lncRNAs are dysregulated in DM and in which pathways they act. Pubmed, Embase, and Gene Expression Omnibus (GEO) repositories were searched to identify studies that investigated lncRNA expression in cases with DM and non-diabetic controls. LncRNAs consistently dysregulated in DM patients were submitted to bioinformatics analysis to retrieve their target genes and identify potentially affected signaling pathways under their regulation. Fifty-three eligible articles were included in this review after the application of the inclusion and exclusion criteria. Six hundred and thirty-eight lncRNAs were differentially expressed between cases and controls in at least one study. Among them, six lncRNAs were consistently dysregulated in patients with DM (Anril, Hotair, Malat1, Miat, Kcnq1ot1, and Meg3) compared to controls. Moreover, these six lncRNAs participate in several metabolism-related pathways, evidencing their importance in DM. This systematic review suggests six lncRNAs are dysregulated in DM, constituting potential biomarkers of this disease.
Collapse
Affiliation(s)
- Cristine Dieter
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Post-Graduate Program in Medical Sciences: Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | - Taís Silveira Assmann
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Post-Graduate Program in Medical Sciences: Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Daisy Crispim
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Post-Graduate Program in Medical Sciences: Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- *Correspondence: Daisy Crispim,
| |
Collapse
|
38
|
Savastio S, Cadario F, D'Alfonso S, Stracuzzi M, Pozzi E, Raviolo S, Rizzollo S, Gigliotti L, Boggio E, Bellomo G, Basagni C, Bona G, Rabbone I, Dianzani U, Prodam F. Vitamin D Supplementation Modulates ICOS+ and ICOS- Regulatory T Cell in Siblings of Children With Type 1 Diabetes. J Clin Endocrinol Metab 2020; 105:5897243. [PMID: 32844222 DOI: 10.1210/clinem/dgaa588] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Vitamin D plays an immunoregulatory activity. The aim of this study was to assess the correlation between blood serum 25(OH)D levels and Th17 and Treg circulating subsets, mainly Treg/inducible costimulatory-positive (ICOS+), which seems to have a protective role in autoimmunity, in children with type 1 diabetes mellitus (T1D) and their healthy siblings (S). The secondary aim was to evaluate the impact of vitamin D supplementation on these subsets. PATIENTS AND METHODS 22 T1D and 33 S were enrolled. Glucose, hemoglobin A1c, 25 OH vitamin D (25[OH]D), T helper type 17 (Th17; CD4+CCR6+), regulatory T cells (Treg; CD4+CD25+Foxp3+), and Treg/ICOS+ cells were evaluated. According to human leukocyte antigen (HLA) haplotypes, subjects were classified as "at risk" (HLA+), "protective haplotypes" (HLA-; "nested controls"), and "undetermined" (HLAUND). T1D and S subjects were supplemented with cholecalciferol 1000 IU/die and evaluated after 6 months. RESULTS Vitamin D insufficiency (74.4%) and deficiency (43%) were frequent. S subjects with 25(OH)D levels <25 nmol/L had Th17, Treg (p < 0.01), and Treg/ICOS+ (P < 0.05) percentages higher than subjects with 25(OH)D >75 nmol/L. Treg/ICOS+ percentages (P < 0.05) were higher in HLA- S subjects compared to percentages observed in S with T1D. At baseline, in S subjects, a decreasing trend in Th17 and Treg/ICOS+ values (P < 0.05) from vitamin D deficiency to sufficiency was observed; 25(OH)D levels were negative predictors of Treg/ICOS+ (R2 = 0.301) and Th17 percentages (R2 = 0.138). After 6 months, supplemented S subjects showed higher 25(OH)D levels (P < 0.0001), and lower Th17 (P < 0.0001) and Treg/ICOS+ (P < 0.05) percentages than at baseline; supplemented T1D patients only had a decrease in Th17 levels (P < 0.05). CONCLUSION Serum 25(OH)D levels seem to affect Th17 and Treg cell subsets in S subjects, consistent with its immunomodulating role. HLA role should be investigated in a larger population.
Collapse
Affiliation(s)
- Silvia Savastio
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
| | - Francesco Cadario
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases, Università del Piemonte Orientale, Novara, Italy
| | - Sandra D'Alfonso
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Marta Stracuzzi
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
| | - Erica Pozzi
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
| | - Silvia Raviolo
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
| | - Stefano Rizzollo
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
| | - Luca Gigliotti
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Elena Boggio
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Giorgio Bellomo
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Chiara Basagni
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Gianni Bona
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Ivana Rabbone
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Umberto Dianzani
- Interdisciplinary Research Center of Autoimmune Diseases, Università del Piemonte Orientale, Novara, Italy
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
- SCDU of Clinical Biochemistry, University Hospital Maggiore della Carità, Novara, Italy
| | - Flavia Prodam
- SCDU of Pediatrics, University Hospital Maggiore della Carità, Novara, Italy
- Interdisciplinary Research Center of Autoimmune Diseases, Università del Piemonte Orientale, Novara, Italy
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
39
|
Pandey S, Dvorakova MC. Future Perspective of Diabetic Animal Models. Endocr Metab Immune Disord Drug Targets 2020; 20:25-38. [PMID: 31241444 PMCID: PMC7360914 DOI: 10.2174/1871530319666190626143832] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/06/2019] [Accepted: 04/17/2019] [Indexed: 12/15/2022]
Abstract
Objective The need of today’s research is to develop successful and reliable diabetic animal models for understanding the disease susceptibility and pathogenesis. Enormous success of animal models had already been acclaimed for identifying key genetic and environmental factors like Idd loci and effects of microorganisms including the gut microbiota. Furthermore, animal models had also helped in identifying many therapeutic targets and strategies for immune-intervention. In spite of a quite success, we have acknowledged that many of the discovered immunotherapies are working on animals and did not have a significant impact on human. Number of animal models were developed in the past to accelerate drug discovery pipeline. However, due to poor initial screening and assessment on inequivalent animal models, the percentage of drug candidates who succeeded during clinical trials was very low. Therefore, it is essential to bridge this gap between pre-clinical research and clinical trial by validating the existing animal models for consistency. Results and Conclusion In this review, we have discussed and evaluated the significance of animal models on behalf of published data on PUBMED. Amongst the most popular diabetic animal models, we have selected six animal models (e.g. BioBreeding rat, “LEW IDDM rat”, “Nonobese Diabetic (NOD) mouse”, “STZ RAT”, “LEPR Mouse” and “Zucker Diabetic Fatty (ZDF) rat” and ranked them as per their published literature on PUBMED. Moreover, the vision and brief imagination for developing an advanced and robust diabetic model of 21st century was discussed with the theme of one mice-one human concept including organs-on-chips.
Collapse
Affiliation(s)
- Shashank Pandey
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Magdalena C Dvorakova
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.,Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|
40
|
Denyer AL, Massey JP, Davison LJ, Ollier WER, Catchpole B, Kennedy LJ. Dog leucocyte antigen (DLA) class II haplotypes and risk of canine diabetes mellitus in specific dog breeds. Canine Med Genet 2020; 7:15. [PMID: 33292601 PMCID: PMC7603736 DOI: 10.1186/s40575-020-00093-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022] Open
Abstract
Background Canine diabetes mellitus (DM) is a common endocrine disease in domestic dogs. A number of pathological mechanisms are thought to contribute to the aetiopathogenesis of relative or absolute insulin deficiency, including immune-mediated destruction of pancreatic beta cells. DM risk varies considerably between different dog breeds, suggesting that genetic factors are involved and contribute susceptibility or protection. Associations of particular dog leucocyte antigen (DLA) class II haplotypes with DM have been identified, but investigations to date have only considered all breeds pooled together. The aim of this study was to analyse an expanded data set so as to identify breed-specific diabetes-associated DLA haplotypes. Methods The 12 most highly represented breeds in the UK Canine Diabetes Register were selected for study. DLA-typing data from 646 diabetic dogs and 912 breed-matched non-diabetic controls were analysed to enable breed-specific analysis of the DLA. Dogs were genotyped for allelic variation at DLA-DRB1, -DQA1, -DQB1 loci using DNA sequence-based typing. Genotypes from all three loci were combined to reveal three-locus DLA class II haplotypes, which were evaluated for statistical associations with DM. This was performed for each breed individually and for all breeds pooled together. Results Five dog breeds were identified as having one or more DLA haplotype associated with DM susceptibility or protection. Four DM-associated haplotypes were identified in the Cocker Spaniel breed, of which one haplotype was shared with Border Terriers. In the three breeds known to be at highest risk of DM included in the study (Samoyed, Tibetan Terrier and Cairn Terrier), no DLA haplotypes were found to be associated with DM. Conclusions Novel DLA associations with DM in specific dog breeds provide further evidence that immune response genes contribute susceptibility to this disease in some cases. It is also apparent that DLA may not be contributing obvious or strong risk for DM in some breeds, including the seven breeds analysed for which no associations were identified.
Collapse
Affiliation(s)
- A L Denyer
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hatfield, UK
| | - J P Massey
- Centre for Integrated Genomic Medical Research, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - L J Davison
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.,Department of Clinical Sciences and Services, Royal Veterinary College, Hatfield, UK
| | - W E R Ollier
- Centre for Integrated Genomic Medical Research, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - B Catchpole
- Department of Pathology and Pathogen Biology, Royal Veterinary College, Hatfield, UK
| | - L J Kennedy
- Centre for Integrated Genomic Medical Research, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
41
|
Al Theyab A, Almutairi T, Al-Suwaidi AM, Bendriss G, McVeigh C, Chaari A. Epigenetic Effects of Gut Metabolites: Exploring the Path of Dietary Prevention of Type 1 Diabetes. Front Nutr 2020; 7:563605. [PMID: 33072796 PMCID: PMC7541812 DOI: 10.3389/fnut.2020.563605] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes (T1D) has increased over the past half century and has now become the second most frequent autoimmune disease in childhood and one of major public health concern worldwide. Evidence suggests that modern lifestyles and rapid environmental changes are driving factors that underlie this increase. The integration of these two factors brings about changes in food intake. This, in turn, alters epigenetic regulations of the genome and intestinal microbiota composition, which may ultimately play a role in pathogenesis of T1D. Recent evidence shows that dysbiosis of the gut microbiota is closely associated with T1D and that a dietary intervention can influence epigenetic changes associated with this disease and may modify gene expression patterns through epigenetic mechanisms. In this review focus on how a diet can shape the gut microbiome, its effect on the epigenome in T1D, and the future of T1D management by microbiome therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Ali Chaari
- Premedical Division, Weill Cornell Medicine Qatar, Doha, Qatar
| |
Collapse
|
42
|
Sirdah MM, Reading NS. Genetic predisposition in type 2 diabetes: A promising approach toward a personalized management of diabetes. Clin Genet 2020; 98:525-547. [PMID: 32385895 DOI: 10.1111/cge.13772] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/04/2020] [Accepted: 05/04/2020] [Indexed: 02/06/2023]
Abstract
Diabetes mellitus, also known simply as diabetes, has been described as a chronic and complex endocrine metabolic disorder that is a leading cause of death across the globe. It is considered a key public health problem worldwide and one of four important non-communicable diseases prioritized for intervention through world health campaigns by various international foundations. Among its four categories, Type 2 diabetes (T2D) is the commonest form of diabetes accounting for over 90% of worldwide cases. Unlike monogenic inherited disorders that are passed on in a simple pattern, T2D is a multifactorial disease with a complex etiology, where a mixture of genetic and environmental factors are strong candidates for the development of the clinical condition and pathology. The genetic factors are believed to be key predisposing determinants in individual susceptibility to T2D. Therefore, identifying the predisposing genetic variants could be a crucial step in T2D management as it may ameliorate the clinical condition and preclude complications. Through an understanding the unique genetic and environmental factors that influence the development of this chronic disease individuals can benefit from personalized approaches to treatment. We searched the literature published in three electronic databases: PubMed, Scopus and ISI Web of Science for the current status of T2D and its associated genetic risk variants and discus promising approaches toward a personalized management of this chronic, non-communicable disorder.
Collapse
Affiliation(s)
- Mahmoud M Sirdah
- Division of Hematology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA.,Biology Department, Al Azhar University-Gaza, Gaza, Palestine
| | - N Scott Reading
- Institute for Clinical and Experimental Pathology, ARUP Laboratories, Salt Lake City, Utah, USA.,Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
43
|
Specific gene expression in type 1 diabetic patients with and without cardiac autonomic neuropathy. Sci Rep 2020; 10:5554. [PMID: 32221364 PMCID: PMC7101413 DOI: 10.1038/s41598-020-62498-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 02/18/2020] [Indexed: 12/26/2022] Open
Abstract
We hypothesized that some molecular pathways might interact to initiate the process of nervous tissue destruction, promoting cardiac autonomic neuropathy (CAN) in the course of diabetes type 1 (T1D). The study group consisted of 60 T1D patients (58.33% women/41.67% men), on standard therapy. The control group consisted of twenty healthy volunteers recruited in accordance with age, gender and body weight. The presence of CAN was documented by the Ewing test method (ProSciCard apparatus). A microarray data analysis was performed using Gene Spring version 13. The microarray results for selected genes were confirmed by real-time PCR (qRT-PCR), using specific TaqMan Gene Expression Assays. Plasma IL-6 content was measured by an enzyme-linked immunosorbent assay (ELISA). The p < 0.05 value was considered as statistically significant. The microarray analysis, confirmed by qRTPCR, showed significant up-regulation of autophagy, quantity of mitochondria, quality regulatory genes (mTOR, GABARAPL2) apoptosis, ER-stress and inflammation (NFKB1, IL1b, IL1R1, SOD1), in T1D when compared to the control group. A significantly higher IL-6 protein level was observed in T1D patients, in comparison to the control group. We concluded that the observed changes in gene expression and activation of intracellular pathways give a coherent picture of the important role of oxidative stress in inflammation and the activation of apoptosis in the pathomechanism of DM. The significance of the inflammatory process, confirmed by the increased level of the inflammation biomarker IL-6 in the pathomechanisms of CAN was shown even in patients with properly treated T1D.
Collapse
|
44
|
Singh GC, Ahmed M, Zaid M, Hasnain S. Biochemical, serological, and genetic aspects related to gene HLA-DQB1 and its association with type 1 diabetes mellitus (T1DM). Mol Genet Genomic Med 2020; 8:e1147. [PMID: 32142224 PMCID: PMC7216801 DOI: 10.1002/mgg3.1147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 10/21/2019] [Accepted: 01/08/2020] [Indexed: 12/23/2022] Open
Abstract
Background Type 1 Diabetes Mellitus (T1DM) is the autoimmune disorder of destruction of β cells of pancreas, creating insulin deficiency condition, which leads to hyperglycemia, polyuria, polydipsia, ketoacidosis, and other metabolic disorder especially in children. Different genetic aspects and environmental factors are involved in pathophysiology of the disease. About 20 genes are associated with this disease in which the most common is the different combination of haplotype DRB1‐DQA1‐DQB1 present at HLA gene. At HLA‐DQB1, there are some SNPs which are associated with T1DM. In T1DM, there are number of biochemical, serological parameters which show some abnormalities leading to some complications. Methods Samples were subjected to all biochemical and serological techniques to get the measurement of concentration of glucose, lipid profile (cholesterol, triglycerides, and HDL and LDL cholesterol), urea, creatinine, albumin, insulin, anti‐insulin antibodies, C‐peptides, and leptin. All these values were compared with controls values and statistical analysis was also done on these values. At molecular level, two primers set which were allele specific at HLA‐DQB1, were used to amplify the SNPs, homozygous and heterozygous conditions were stated. Results PCR results for the studied population showed that most of samples have heterozygous condition for these SNPs of this allele specific region on HLA‐DQB1. Very few of them have homozygous state for it. Even in the control sample have the same conditions. Conclusion In Pakistan, there is dire need of studies about SNPs and haplotypes related to HLA‐DQB1 which show association with T1DM.
Collapse
Affiliation(s)
- Gur Charn Singh
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan
| | - Mehboob Ahmed
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan
| | - Muhammad Zaid
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan.,Department of Life Sciences, School of Science, University of Management and Technology, Lahore, Pakistan
| | - Shahida Hasnain
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
45
|
Floris M, Cano A, Porru L, Addis R, Cambedda A, Idda ML, Steri M, Ventura C, Maioli M. Direct-to-Consumer Nutrigenetics Testing: An Overview. Nutrients 2020; 12:566. [PMID: 32098227 PMCID: PMC7071525 DOI: 10.3390/nu12020566] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 01/22/2023] Open
Abstract
At present, specialized companies offering genetic testing services without the involvement of clinicians are growing; this development is a direct consequence of the significant decrease in genotyping and sequencing costs. Online companies offer predictions about the risk of developing complex diseases during one's life course, and they offer suggestions for personal lifestyle. Several companies have been created that provide nutrigenetics services; these companies suggest dietary indications-a central issue in the prevention and etiopathogenesis of specific diseases-based on one's personal genetic background. Dietary patterns are defined on the basis of a limited set of genetic markers. In this article, we analyze the online nutrigenetics services offered by 45 companies worldwide, to obtain an overall picture of the costs, the types of nutritional traits considered and the level of scientific precision of the services proposed. Our analysis clearly highlights the need for specific guidelines, in order to ensure a set of minimum quality standards for the nutrigenetics services offered to the customer.
Collapse
Affiliation(s)
- Matteo Floris
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari, Italy; (A.C.); (L.P.); (A.C.)
- Institute for Genetic and Biomedical Research, National Research Council, traversa La Crucca 3, 07100 Sassari, Italy;
| | - Antonella Cano
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari, Italy; (A.C.); (L.P.); (A.C.)
| | - Laura Porru
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari, Italy; (A.C.); (L.P.); (A.C.)
| | - Roberta Addis
- Department of Chemistry and Pharmacy, University of Sassari, Via Muroni 23/A, 07100 Sassari, Italy;
| | - Antonio Cambedda
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari, Italy; (A.C.); (L.P.); (A.C.)
| | - Maria Laura Idda
- Institute for Genetic and Biomedical Research, National Research Council, traversa La Crucca 3, 07100 Sassari, Italy;
| | - Maristella Steri
- Institute for Genetic and Biomedical Research, National Research Council, SS 554 Km 4,500, Monserrato, 09042 Cagliari, Italy;
| | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems–Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy;
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari, Italy; (A.C.); (L.P.); (A.C.)
- Institute for Genetic and Biomedical Research, National Research Council, SS 554 Km 4,500, Monserrato, 09042 Cagliari, Italy;
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems–Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy;
- Center for Developmental Biology and Reprogramming-CEDEBIOR, Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari, Italy
| |
Collapse
|
46
|
Cerna M. Epigenetic Regulation in Etiology of Type 1 Diabetes Mellitus. Int J Mol Sci 2019; 21:ijms21010036. [PMID: 31861649 PMCID: PMC6981658 DOI: 10.3390/ijms21010036] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is caused by an autoimmune destruction of the pancreatic β-cells, a process in which autoreactive T cells play a pivotal role, and it is characterized by islet autoantibodies. Consequent hyperglycemia is requiring lifelong insulin replacement therapy. T1DM is caused by the interaction of multiple environmental and genetic factors. The integrations of environments and genes occur via epigenetic regulations of the genome, which allow adaptation of organism to changing life conditions by alternation of gene expression. T1DM has increased several-fold over the past half century. Such a short time indicates involvement of environment factors and excludes genetic changes. This review summarizes the most current knowledge of epigenetic changes in that process leading to autoimmune diabetes mellitus.
Collapse
Affiliation(s)
- Marie Cerna
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Ruska 87, 100 00 Prague 10, Czech Republic
| |
Collapse
|
47
|
Abstract
Type 1 diabetes mellitus (T1DM) is characterized by autoimmune destruction of pancreatic beta-cells in genetically predisposed individuals, eventually resulting in severe insulin deficiency. It is the most common form of diabetes in children and adolescents. Genetic susceptibility plays a crucial role in development of T1DM. The human leukocyte antigen complex plays a key role in the pathogenesis of T1DM. Furthermore, genome-wide association studies and linkage analysis have recently made a significant contribution to current knowledge relative to the impact of genetics on T1DM development and progression. This review focuses on current knowledge of genetics as a pathogenesis for T1DM. It also discusses mechanisms by which genes influence the risk of developing T1DM as well as the clinical and research applications of genetic risk scores in T1DM.
Collapse
Affiliation(s)
- Hae Sang Lee
- Department of Pediatrics, Ajou University Hospital, Ajou University School of Medicine, Suwon, Korea,Address for correspondence: Hae Sang Lee, MD, PhD Department of Pediatrics, Ajou University Hospital, Ajou University School of Medicine, 164 World cupro, Yeongtong-gu, Suwon 16499, Korea Tel: +82-31-219-5166 Fax: +82-31-219-5169 E-mail:
| | - Jin Soon Hwang
- Department of Pediatrics, Ajou University Hospital, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
48
|
Blanter M, Sork H, Tuomela S, Flodström-Tullberg M. Genetic and Environmental Interaction in Type 1 Diabetes: a Relationship Between Genetic Risk Alleles and Molecular Traits of Enterovirus Infection? Curr Diab Rep 2019; 19:82. [PMID: 31401790 PMCID: PMC6689284 DOI: 10.1007/s11892-019-1192-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW We provide an overview of the current knowledge regarding the natural history of human type 1 diabetes (T1D) and the documented associations between virus infections (in particular the enteroviruses) and disease development. We review studies that examine whether T1D-specific risk alleles in genes involved in the function of the immune system can alter susceptibility to virus infections or affect the magnitude of the host antiviral response. We also highlight where the major gaps in our knowledge exist and consider possible implications that new insights gained from the discussed gene-environment interaction studies may bring. RECENT FINDINGS A commonality between several of the studied T1D risk variants studied is their role in modulating the host immune response to viral infection. Generally, little support exists indicating that the risk variants increase susceptibility to infection and moreover, they usually appear to predispose the immune system towards a hyper-reactive state, decrease the risk of infection, and/or favor the establishment of viral persistence. In conclusion, although the current number of studies is limited, this type of research can provide important insights into the mechanisms that are central to disease pathogenesis and further describe how genetic and environmental factors jointly influence the risk of T1D development. The latter may provide genetic markers that could be used for patient stratification and for the selection of method(s) for disease prevention.
Collapse
Affiliation(s)
- Marfa Blanter
- 0000 0000 9241 5705grid.24381.3cCenter for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- 0000 0001 0668 7884grid.5596.fLaboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven, EU Belgium
| | - Helena Sork
- 0000 0000 9241 5705grid.24381.3cCenter for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Soile Tuomela
- 0000 0000 9241 5705grid.24381.3cCenter for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Malin Flodström-Tullberg
- 0000 0000 9241 5705grid.24381.3cCenter for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|