1551
|
Rekers NV, von Herrath MG, Wesley JD. Immunotherapies and immune biomarkers in Type 1 diabetes: A partnership for success. Clin Immunol 2015; 161:37-43. [PMID: 26122172 DOI: 10.1016/j.clim.2015.05.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/13/2015] [Accepted: 05/17/2015] [Indexed: 12/16/2022]
Abstract
The standard of care (SoC) for Type 1 diabetes (T1D) today is much the same as it was in the early 1920s, simply with more insulin options-fast-acting, slow-acting, injectable, and inhalable insulins. However, these well-tolerated treatments only manage the symptoms and complications, but do nothing to halt the underlying immune response. There is an unmet need for better treatment options for T1D that address all aspects of the disease. For decades, we have successfully treated T1D in preclinical animal models with immune-modifying therapies that have not demonstrated comparable efficacy in humans. The path to bringing such options to the clinic will depend on the implementation and standard inclusion of biomarkers of immune and therapeutic efficacy in T1D clinical trials, and dictate if we can create a new SoC that treats the underlying autoimmunity as well as the symptoms it causes.
Collapse
Affiliation(s)
- Niels V Rekers
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, WA, USA; Pacific Northwest Diabetes Research Institute, Seattle, WA, USA
| | | | - Johnna D Wesley
- Type 1 Diabetes R&D Center, Novo Nordisk Inc., Seattle, WA, USA.
| |
Collapse
|
1552
|
Nyitray CE, Chang R, Faleo G, Lance KD, Bernards DA, Tang Q, Desai T. Polycaprolactone Thin-Film Micro- and Nanoporous Cell-Encapsulation Devices. ACS NANO 2015; 9:5675-82. [PMID: 25950860 PMCID: PMC4628825 DOI: 10.1021/acsnano.5b00679] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Cell-encapsulating devices can play an important role in advancing the types of tissue available for transplantation and further improving transplant success rates. To have an effective device, encapsulated cells must remain viable, respond to external stimulus, and be protected from immune responses, and the device itself must elicit a minimal foreign body response. To address these challenges, we developed a micro- and a nanoporous thin-film cell encapsulation device from polycaprolactone (PCL), a material previously used in FDA-approved biomedical devices. The thin-film device construct allows long-term bioluminescent transfer imaging, which can be used for monitoring cell viability and device tracking. The ability to tune the microporous and nanoporous membrane allows selective protection from immune cell invasion and cytokine-mediated cell death in vitro, all while maintaining typical cell function, as demonstrated by encapsulated cells' insulin production in response to glucose stimulation. To demonstrate the ability to track, visualize, and monitor the viability of cells encapsulated in implanted thin-film devices, we encapsulated and implanted luciferase-positive MIN6 cells in allogeneic mouse models for up to 90 days. Lack of foreign body response in combination with rapid neovascularization around the device shows promise in using this technology for cell encapsulation. These devices can help elucidate the metrics required for cell encapsulation success and direct future immune-isolation therapies.
Collapse
Affiliation(s)
- Crystal E. Nyitray
- Program in Chemistry and Chemical Biology, University of California, San Francisco, 1700 4th Street, Byers Hall, Box 2520, San Francisco, California 94158, United States
| | - Ryan Chang
- UCB/UCSF Joint Program in Bioengineering, University of California, San Francisco, 1700 4th Street, Byers Hall, Box 2520, San Francisco, California 94158, United States
| | - Gaetano Faleo
- Department of Surgery, University of California, San Francisco, 513 Parnassus Avenue HSE520 Box 0780, San Francisco, California 94143, United States
| | - Kevin D. Lance
- UCB/UCSF Joint Program in Bioengineering, University of California, San Francisco, 1700 4th Street, Byers Hall, Box 2520, San Francisco, California 94158, United States
| | - Daniel A. Bernards
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1700 4th Street, Byers Hall, Box 2520, San Francisco, California 94158, United States
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, 513 Parnassus Avenue HSE520 Box 0780, San Francisco, California 94143, United States
| | - TejalA Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, 1700 4th Street, Byers Hall, Box 2520, San Francisco, California 94158, United States
| |
Collapse
|
1553
|
Cechin SR, Lopez-Ocejo O, Karpinsky-Semper D, Buchwald P. Biphasic decline of β-cell function with age in euglycemic nonobese diabetic mice parallels diabetes onset. IUBMB Life 2015; 67:634-44. [PMID: 26099053 DOI: 10.1002/iub.1391] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 05/27/2015] [Indexed: 01/10/2023]
Abstract
A gradual decline in insulin response is known to precede the onset of type 1 diabetes (T1D). To track age-related changes in the β-cell function of nonobese diabetic (NOD) mice, the most commonly used animal model for T1D, and to establish differences between those who do and do not become hyperglycemic, we performed a long-term longitudinal oral glucose tolerance test (OGTT) study (10-42 weeks) in combination with immunofluorescence imaging of islet morphology and cell proliferation. We observed a clear biphasic decline in insulin secretion (AUC0-30 min ) even in euglycemic animals. A first phase (10-28 weeks) consisted of a relatively rapid decline and paralleled diabetes development in the same cohort of animals. This was followed by a second phase (29-42 weeks) during which insulin secretion declined much slower while no additional animals became diabetic. Blood glucose profiles showed a corresponding, but less pronounced change: the area under the concentration curve (AUC0-150 min ) increased with age, and fit with a bilinear model indicated a rate-change in the trendline around 28 weeks. In control NOD scids, no such changes were observed. Islet morphology also changed with age as islets become surrounded by mononuclear infiltrates, and, in all mice, islets with immune cell infiltration around them showed increased β-cell proliferation. In conclusion, insulin secretion declines in a biphasic manner in all NOD mice. This trend, as well as increased β-cell proliferation, is present even in the NODs that never become diabetic, whereas, it is absent in control NOD scid mice.
Collapse
Affiliation(s)
- Sirlene R Cechin
- Diabetes Research Institute, Miller School of Medicine, University of Miami, FL, USA
| | - Omar Lopez-Ocejo
- Diabetes Research Institute, Miller School of Medicine, University of Miami, FL, USA
| | | | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, FL, USA.,Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, FL, USA
| |
Collapse
|
1554
|
Abstract
Type 1 and type 2 diabetes are growing public health problems. Despite having different pathophysiologies, both diseases are associated with defects in immune regulation. Invariant natural killer T (iNKT) cells are innate-like T cells that recognize glycolipids presented by CD1d. These cells not only play a key role in the defense against pathogens, but also exert potent immunoregulatory functions. The regulatory role of iNKT cells in the prevention of type 1 diabetes has been demonstrated in murine models and analyzed in diabetic patients. The decreased frequency of iNKT cells in non-obese diabetic mice initially suggested the regulatory role of this cell subset. Increasing the frequency or the activation of iNKT cells with agonists protects non-obese diabetic mice from the development of diabetes. Several mechanisms mediate iNKT regulatory functions. They can rapidly produce immunoregulatory cytokines, interleukin (IL)-4 and IL-10. They induce tolerogenic dendritic cells, thereby inducing the anergy of autoreactive anti-islet T cells and increasing the frequency of T regulatory cells (Treg cells). Synthetic agonists are able to activate iNKT cells and represent potential therapeutic treatment in order to prevent type 1 diabetes. Growing evidence points to a role of immune system in glucose intolerance and type 2 diabetes. iNKT cells are resident cells of adipose tissue and their local and systemic frequencies are reduced in obese patients, suggesting their involvement in local and systemic inflammation during obesity. With the discovery of potential continuity between type 1 and type 2 diabetes in some patients, the role of iNKT cells in these diseases deserves further investigation.
Collapse
Affiliation(s)
| | | | - Agnes Lehuen
- Laboratory "Immunology of Diabetes" U1016 INSERM Institut Cochin; CNRS UMR8104; Laboratoire d'Excellence INFLAMEX, Université Paris Descartes, Sorbonne Paris Cité; DHU Authors, Hôpital Cochin, 75014, Paris, France
| |
Collapse
|
1555
|
Atkinson MA, von Herrath M, Powers AC, Clare-Salzler M. Current concepts on the pathogenesis of type 1 diabetes--considerations for attempts to prevent and reverse the disease. Diabetes Care 2015; 38:979-88. [PMID: 25998290 PMCID: PMC4439528 DOI: 10.2337/dc15-0144] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Mark A Atkinson
- Department of Pathology, University of Florida, Gainesville, FL Department of Pediatrics, University of Florida, Gainesville, FL
| | - Matthias von Herrath
- La Jolla Institute for Allergy and Immunology, San Diego, CA Novo Nordisk R&D Center, Seattle, WA
| | - Alvin C Powers
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University, Nashville, TN Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN VA Tennessee Valley Healthcare System, Nashville, TN
| | | |
Collapse
|
1556
|
Jacobsen R, Hypponen E, Sørensen TIA, Vaag AA, Heitmann BL. Gestational and Early Infancy Exposure to Margarine Fortified with Vitamin D through a National Danish Programme and the Risk of Type 1 Diabetes: The D-Tect Study. PLoS One 2015; 10:e0128631. [PMID: 26030061 PMCID: PMC4452099 DOI: 10.1371/journal.pone.0128631] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 04/30/2015] [Indexed: 12/02/2022] Open
Abstract
The objective of the study was to assess whether gestational and early infancy exposure to low dose vitamin D from a mandatory margarine fortification programme in Denmark influenced the risk of developing type 1 diabetes (T1D) before age of 15 years. The study population included all individuals born in Denmark from 1983 to 1988 and consisted of 331,623 individuals. The 1st of June 1985, which was the date of issue of the new ministerial order cancelling mandatory fortification of margarine with vitamin D in Denmark, served as a reference point separating the studied population into various exposure groups. We further modelled birth cohort effects in children developing T1D as a linear spline, and compared the slopes between the birth cohorts with various prenatal and infancy exposures to vitamin D fortification. In total, 886 (0.26%) individuals developed T1D before the age of 15 years. The beta coefficients (95% CI), or slopes, for linear birth cohort effect in log Hazard Ratio (HR) per one month of birth in individuals born during the periods of gestational exposure, wash-out, and non-exposure were: 0.010 (-0.002/0.021), -0.010 (-0.035/0.018), and 0.008 (- 0.017/0.032), respectively. The beta coefficients (95% CI) for individuals born during the periods of first postnatal year exposure, wash-out, and non-exposure were: 0.007 (-0.016/0.030), 0.006 (-0.004/0.016), and 0.007 (-0.002/0.016), respectively. In conclusion, we found no evidence to support that exposure to low dose vitamin D from the Danish mandatory margarine fortification regimen during gestational and first postnatal year of life changed the risk of developing T1D before the age of 15 years.
Collapse
Affiliation(s)
- Ramune Jacobsen
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospitals, The Capital Region, Frederiksberg, Denmark
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospitals, The Capital Region, Frederiksberg, Denmark
- * E-mail:
| | - Elina Hypponen
- School of Population Health, University of South Australia, Adelaide, Australia
| | - Thorkild I. A. Sørensen
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospitals, The Capital Region, Frederiksberg, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Allan A. Vaag
- Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Berit L. Heitmann
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospitals, The Capital Region, Frederiksberg, Denmark
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospitals, The Capital Region, Frederiksberg, Denmark
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
- Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, University of Sydney, Sydney, Australia
| |
Collapse
|
1557
|
Transcriptional regulation of chemokine genes: a link to pancreatic islet inflammation? Biomolecules 2015; 5:1020-34. [PMID: 26018641 PMCID: PMC4496708 DOI: 10.3390/biom5021020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 05/12/2015] [Indexed: 12/18/2022] Open
Abstract
Enhanced expression of chemotactic cytokines (aka chemokines) within pancreatic islets likely contributes to islet inflammation by regulating the recruitment and activation of various leukocyte populations, including macrophages, neutrophils, and T-lymphocytes. Because of the powerful actions of these chemokines, precise transcriptional control is required. In this review, we highlight what is known about the signals and mechanisms that govern the transcription of genes encoding specific chemokine proteins in pancreatic islet β-cells, which include contributions from the NF-κB and STAT1 pathways. We further discuss increased chemokine expression in pancreatic islets during autoimmune-mediated and obesity-related development of diabetes.
Collapse
|
1558
|
Abstract
Diabetes is one of the most common chronic medical disorders in children. The management of diabetes remains a substantial burden on children with diabetes and their families, despite improvements in treatment and rates of morbidity and mortality. Although most children with diabetes have type 1 diabetes, the increasing recognition of type 2 diabetes and genetic forms of diabetes in the paediatric population has important treatment implications. Diabetes therapy focuses strongly on targets for good metabolic control to reduce the risk of long-term complications. A parallel goal is to minimise short-term complications of hypoglycaemia and diabetic ketoacidosis. Technology offers opportunity for improvement in care, but has not yet fully lived up to its potential. New insights into the pathogenesis of diabetes and the development of new therapies have led to clinical trials aimed at the prevention of diabetes.
Collapse
Affiliation(s)
- Fergus J Cameron
- Centre for Hormone Research, Department of Endocrinology and Diabetes, The Royal Children's Hospital, Murdoch Children's Research Institute and Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Diane K Wherrett
- Division of Endocrinology, Department of Paediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
1559
|
Mukherjee G, Chaparro RJ, Schloss J, Smith C, Bando CD, DiLorenzo TP. Glucagon-reactive islet-infiltrating CD8 T cells in NOD mice. Immunology 2015; 144:631-40. [PMID: 25333865 DOI: 10.1111/imm.12415] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 10/13/2014] [Accepted: 10/17/2014] [Indexed: 01/13/2023] Open
Abstract
Type 1 diabetes is characterized by T-cell-mediated destruction of the insulin-producing β cells in pancreatic islets. A number of islet antigens recognized by CD8 T cells that contribute to disease pathogenesis in non-obese diabetic (NOD) mice have been identified; however, the antigenic specificities of the majority of the islet-infiltrating cells have yet to be determined. The primary goal of the current study was to identify candidate antigens based on the level and specificity of expression of their genes in mouse islets and in the mouse β cell line MIN6. Peptides derived from the candidates were selected based on their predicted ability to bind H-2K(d) and were examined for recognition by islet-infiltrating T cells from NOD mice. Several proteins, including those encoded by Abcc8, Atp2a2, Pcsk2, Peg3 and Scg2, were validated as antigens in this way. Interestingly, islet-infiltrating T cells were also found to recognize peptides derived from proglucagon, whose expression in pancreatic islets is associated with α cells, which are not usually implicated in type 1 diabetes pathogenesis. However, type 1 diabetes patients have been reported to have serum autoantibodies to glucagon, and NOD mouse studies have shown a decrease in α cell mass during disease pathogenesis. Our finding of islet-infiltrating glucagon-specific T cells is consistent with these reports and suggests the possibility of α cell involvement in development and progression of disease.
Collapse
Affiliation(s)
- Gayatri Mukherjee
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | | | | | | | |
Collapse
|
1560
|
Krogvold L, Edwin B, Buanes T, Frisk G, Skog O, Anagandula M, Korsgren O, Undlien D, Eike MC, Richardson SJ, Leete P, Morgan NG, Oikarinen S, Oikarinen M, Laiho JE, Hyöty H, Ludvigsson J, Hanssen KF, Dahl-Jørgensen K. Detection of a low-grade enteroviral infection in the islets of langerhans of living patients newly diagnosed with type 1 diabetes. Diabetes 2015; 64:1682-1687. [PMID: 25422108 DOI: 10.2337/db14-1370] [Citation(s) in RCA: 234] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 11/18/2014] [Indexed: 02/05/2023]
Abstract
The Diabetes Virus Detection study (DiViD) is the first to examine fresh pancreatic tissue at the diagnosis of type 1 diabetes for the presence of viruses. Minimal pancreatic tail resection was performed 3-9 weeks after onset of type 1 diabetes in six adult patients (age 24-35 years). The presence of enteroviral capsid protein 1 (VP1) and the expression of class I HLA were investigated by immunohistochemistry. Enterovirus RNA was analyzed from isolated pancreatic islets and from fresh-frozen whole pancreatic tissue using PCR and sequencing. Nondiabetic organ donors served as controls. VP1 was detected in the islets of all type 1 diabetic patients (two of nine controls). Hyperexpression of class I HLA molecules was found in the islets of all patients (one of nine controls). Enterovirus-specific RNA sequences were detected in four of six patients (zero of six controls). The results were confirmed in various laboratories. Only 1.7% of the islets contained VP1(+) cells, and the amount of enterovirus RNA was low. The results provide evidence for the presence of enterovirus in pancreatic islets of type 1 diabetic patients, which is consistent with the possibility that a low-grade enteroviral infection in the pancreatic islets contributes to disease progression in humans.
Collapse
Affiliation(s)
- Lars Krogvold
- Paediatric Department, Oslo University Hospital, Oslo, Norway
| | - Bjørn Edwin
- Intervention Centre and Department of Surgery, Oslo University Hospital, Oslo, Norway Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trond Buanes
- Faculty of Medicine, University of Oslo, Oslo, Norway Department of Surgery, Oslo University Hospital, Oslo, Norway
| | - Gun Frisk
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Oskar Skog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Mahesh Anagandula
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Dag Undlien
- Faculty of Medicine, University of Oslo, Oslo, Norway Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Morten C Eike
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Sarah J Richardson
- Institute of Biomedical & Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Pia Leete
- Institute of Biomedical & Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Noel G Morgan
- Institute of Biomedical & Clinical Science, University of Exeter Medical School, Exeter, U.K
| | - Sami Oikarinen
- Department of Virology, School of Medicine, University of Tampere, Tampere, Finland
| | - Maarit Oikarinen
- Department of Virology, School of Medicine, University of Tampere, Tampere, Finland
| | - Jutta E Laiho
- Department of Virology, School of Medicine, University of Tampere, Tampere, Finland
| | - Heikki Hyöty
- Department of Virology, School of Medicine, University of Tampere, Tampere, Finland Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| | - Johnny Ludvigsson
- Division of Paediatrics, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Kristian F Hanssen
- Faculty of Medicine, University of Oslo, Oslo, Norway Department of Endocrinology, Oslo University Hospital, Oslo, Norway
| | - Knut Dahl-Jørgensen
- Paediatric Department, Oslo University Hospital, Oslo, Norway Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
1561
|
Abstract
Immunotherapies for type 1 diabetes mellitus (T1DM) have been the focus of intense basic and clinical research over the past few decades. Restoring β-cell function is the ultimate goal of intervention trials that target the immune system in T1DM. In an attempt to achieve this aim, different combination therapies have been proposed over the past few years that are based on treatments tackling the various mechanisms involved in the destruction of β cells. The results of clinical trials have not matched expectations based on the positive results from preclinical studies. The heterogeneity of T1DM might explain the negative results obtained, but previous trials have not addressed this issue. However, novel promising combination therapies are being developed, including those that couple immunomodulators with drugs that stimulate β-cell regeneration in order to restore normoglycaemia. This strategy is an encouraging one to pursue the goal of finding a cure for T1DM. This Review summarizes the available data about combination immunotherapies in T1DM, particularly addressing their clinical importance. The available data supporting the use of registered drugs, such as proton pump inhibitors and incretin-based agents, that have been shown to induce β-cell regeneration will also be discussed.
Collapse
Affiliation(s)
- Paolo Pozzilli
- Department of Endocrinology and Diabetes, University Campus Bio-Medico, Via Álvaro del Portillo 21, Rome 00128, Italy
| | - Ernesto Maddaloni
- Department of Endocrinology and Diabetes, University Campus Bio-Medico, Via Álvaro del Portillo 21, Rome 00128, Italy
| | - Raffaella Buzzetti
- Department of Experimental Medicine, "Sapienza" University, Viale Regina Elena 324, Rome 00161 Italy
| |
Collapse
|
1562
|
Smolina K, Wotton CJ, Goldacre MJ. Risk of dementia in patients hospitalised with type 1 and type 2 diabetes in England, 1998-2011: a retrospective national record linkage cohort study. Diabetologia 2015; 58:942-50. [PMID: 25673256 DOI: 10.1007/s00125-015-3515-x] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 01/15/2015] [Indexed: 12/23/2022]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes increases the risk of subsequent dementia. Our objective was to determine whether a similar risk of subsequent dementia is associated with type 1 diabetes in a large defined population. METHODS This retrospective cohort study examined national administrative record-linked statistical data on hospital care and mortality in England, 1998-2011. Cohorts of people admitted to hospital when aged 30 or over were constructed: 343,062 people with type 1 diabetes; 1,855,141 people with type 2 diabetes; and a reference cohort. Results were expressed as rate ratios (RR) comparing each diabetes cohort with the control cohort. RESULTS The overall RR for dementia in people admitted to hospital with type 1 diabetes was 1.65 (95% CI 1.61, 1.68), and for people admitted to hospital with type 2 diabetes was 1.37 (1.35, 1.38). Young age at admission for diabetes appeared to confer a greater rate of subsequent dementia; the RR for dementia in people admitted to hospital with type 1 diabetes aged 30-39 years was 7.10 (4.65, 10.6), which reduced to 4.40 (3.55, 5.40) in those aged 40-49 at admission, and further reduced with increasing age to 1.16 (1.11, 1.20) in those aged 80 or over at admission. A similar pattern was seen with type 2 diabetes. CONCLUSIONS/INTERPRETATION Type 1 diabetes, as well as type 2 diabetes, may be associated with an elevated risk of subsequent dementia. The risk of dementia varies with age at admission to hospital with diabetes, and appears to be much greater in the young.
Collapse
Affiliation(s)
- Kate Smolina
- Centre for Health Services and Policy Research, School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|
1563
|
An artificial pancreas for automated blood glucose control in patients with Type 1 diabetes. Ther Deliv 2015; 6:609-19. [DOI: 10.4155/tde.15.12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Automated glucose control in patients with Type 1 diabetes is much-coveted by patients, relatives and healthcare professionals. It is the expectation that a system for automated control, also know as an artificial pancreas, will improve glucose control, reduce the risk of diabetes complications and markedly improve patient quality of life. An artificial pancreas consists of portable devices for glucose sensing and insulin delivery which are controlled by an algorithm residing on a computer. The technology is still under development and currently no artificial pancreas is commercially available. This review gives an introduction to recent progress, challenges and future prospects within the field of artificial pancreas research.
Collapse
|
1564
|
Abstract
Patients with type 1 diabetes (T1D) suffer excess mortality from cardiovascular disease (CVD) that has persisted despite substantial reductions in microvascular complications. Although T1D and type 2 diabetes (T2D) are etiologically distinct, it has generally been assumed that CVD in T1D is "the same disease" as that found in T2D. Here, we review the most recent epidemiological and clinical studies on heart disease in T1D, highlighting differences between CVD in T1D and T2D. In addition, we discuss experimental and clinical evidence for a post-myocardial infarction (MI) autoimmune heart syndrome in T1D, including the development of diagnostic assays which we believe can, for the first time, differentiate between heart disease in T1D and T2D. We postulate that a clinically unrecognized form of chronic myocardial inflammation ("myocarditis") triggered by MI contributes to the poor CVD outcomes in T1D. These findings provide a conceptual shift in our understanding of CVD in T1D and have important diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Myra A Lipes
- Joslin Diabetes Center, Harvard Medical School, 1 Joslin Place, Rm. 373, Boston, MA, 02215, USA,
| | | |
Collapse
|
1565
|
Kuehl MN, Rodriguez H, Burkhardt BR, Alman AC. Tumor Necrosis Factor-α, Matrix-Metalloproteinases 8 and 9 Levels in the Saliva Are Associated with Increased Hemoglobin A1c in Type 1 Diabetes Subjects. PLoS One 2015; 10:e0125320. [PMID: 25915398 PMCID: PMC4411091 DOI: 10.1371/journal.pone.0125320] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/14/2015] [Indexed: 11/19/2022] Open
Abstract
Background Type 1 diabetes (T1D) is an autoimmune disease resulting in the targeted destruction of pancreatic β-cells and permanent loss of insulin production. Proper glucose management results in better clinical outcomes for T1D and provides a strong rationale to identify non-invasive biomarkers indicative or predictive of glycemic control. Therefore, we investigated the association of salivary inflammation with HbA1c in a T1D cohort. Methods Unstimulated saliva was collected from 144 subjects with T1D at the USF Diabetes Center. BMI, duration of diabetes, and HbA1c were recorded during clinical visit. Levels of interleukin (IL)-1β, -6, -8, -10, IFN-γ, TNF-α, MMP-3, -8, and -9 were measured using multiplexing immunoassay analysis. To account for smoking status, salivary cotinine levels were also determined. Results Multiple linear (HbA1c) and logistic (self-reported gingival condition) regression analyses were performed to examine the relationships between the Principal Component Analysis (PCA) components and HbA1c and gingival condition (adjusted for age, duration of diabetes, BMI, and sex; model for HbA1c also adjusted for gingival condition and model for gingival condition also adjusted for HbA1c). PCA components 1 (MMP-8 and MMP-9) and 3 (TNF-α) were significantly associated with HbA1c (β = 0.28 ±0.14, p = 0.045; β = 0.31 ±0.14, p = 0.029), while PCA component 2 (IL-6, IL-1β, and IL-8) was significantly associated with gingival condition (OR 1.60 95% CI 1.09–2.34, p = 0.016). In general, increased salivary inflammatory burden is associated with decreased glycemic control and self-reported gingival condition. Conclusions The saliva may represent a useful reservoir of novel noninvasive inflammatory biomarkers predictive of the progression and control of T1D.
Collapse
Affiliation(s)
- Melanie N. Kuehl
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, Florida, the United States of America
| | - Henry Rodriguez
- Diabetes Center, College of Medicine, University of South Florida, Tampa, Florida, the United States of America
| | - Brant R. Burkhardt
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, Florida, the United States of America
- Diabetes Center, College of Medicine, University of South Florida, Tampa, Florida, the United States of America
- * E-mail:
| | - Amy C. Alman
- Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| |
Collapse
|
1566
|
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by destruction of insulin-producing β cells in the pancreas. The incidence of T1D is increasing dramatically, and the prevalence has doubled in the last 2 decades, further increasing the morbidity and mortality associated with the disease. T1D is now predictable with the measurement of antibodies directed against β cell proteins. Islet autoantibodies (IAs) are detectable from the peripheral blood months to years before clinical diagnosis. With the presence of two or more antibodies, the risk for developing T1D is nearly 100 % given enough time. Targeted screening for T1D risk has been carried out in first-degree relatives and those with a significant genetic risk. However, more than 85 % of individuals who are diagnosed with T1D do not have a family history. In light of the predictability of T1D and recent advances in IA measurement, general population screening is on the horizon. We provide an overview of the history of general population screening and discuss the rationale for and arguments against screening the general population for T1D risk.
Collapse
Affiliation(s)
- Kimber M Simmons
- Pediatric Endocrinology and Diabetes Fellow, Children's Hospital Colorado, Aurora, Colorado, US
| | - Aaron W Michels
- Assistant Professor of Pediatrics & Medicine, Barbara Davis Center for Childhood Diabetes, University of Colorado, Denver, US
| |
Collapse
|
1567
|
Abstract
BACKGROUND Celiac disease (CD) is a small-intestinal inflammatory disease that is triggered by the ingestion of the storage proteins (gluten) of wheat, barley and rye. KEY MESSAGES Endocrine autoimmunity is prevalent in patients with CD and their relatives. The genes that predispose to endocrine autoimmune diseases, e.g. type 1 diabetes, autoimmune thyroid diseases, and Addison's disease, i.e. DR3-DQ2 and DR4-DQ8, are also the major genetic determinants of CD, which is the best understood HLA-linked disease. Thus, up to 30% of first-degree relatives both of patients with CD and/or endocrine autoimmunity are affected by the other disease. In CD, certain gluten proteins bind with high affinity to HLA-DQ2 or -DQ8 in the small-intestinal mucosa, to activate gluten-specific T cells which are instrumental in the destruction of the resorptive villi. Here, the autoantigen tissue transglutaminase increases the T cell response by generating deamidated gluten peptides that bind more strongly to DQ2 or DQ8. Classical symptoms such as diarrhea and consequences of malabsorption like anemia and osteoporosis are often absent in patients with (screening-detected) CD, but this absence does not significantly affect these patients' incidence of endocrine autoimmunity. Moreover, once autoimmunity is established, a gluten-free diet is not able to induce remission. However, ongoing studies attempt to address how far a gluten-free diet may prevent or retard the development of CD and endocrine autoimmunity in children at risk. CONCLUSIONS The close relationship between CD and endocrine autoimmunity warrants a broader immune genetic and endocrine screening of CD patients and their relatives.
Collapse
|
1568
|
Low L, Law JP, Hodson J, McAlpine R, O'Colmain U, MacEwen C. Impact of socioeconomic deprivation on the development of diabetic retinopathy: a population-based, cross-sectional and longitudinal study over 12 years. BMJ Open 2015; 5:e007290. [PMID: 25877277 PMCID: PMC4401835 DOI: 10.1136/bmjopen-2014-007290] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE To study the association between socioeconomic deprivation and prevalence of diabetic retinopathy (DR). DESIGN Population-based, cross-sectional observational study and retrospective longitudinal analysis over 12 years. SETTING Primary care, East of Scotland. METHODS Outcome data from DR screening examinations (digital retinal photography) were collected from the Scottish regional diabetes electronic record from inception of database to December 2012. The overall Scottish Index of Multiple Deprivation (SIMD) 2012 score for each patient was obtained using their residential postcode. Multiple binary logistic regression was used to analyse the relationship between overall SIMD score and prevalence of DR, adjusting for other variables: age, gender, glycated haemoglobin, cholesterol levels and duration of disease. PRIMARY OUTCOME Any retinopathy (R1 and above) in either eye. RESULTS A total of 1861 patients with type 1 diabetes mellitus (DM) and 18,197 patients with type 2 DM were included in the study. Prevalence of DR in type 1 and type 2 DM were 56.3% and 25.5%, respectively. Increased prevalence of DR in type 1 DM was associated with higher overall SIMD score (p=0.002), with an OR for the most deprived relative to the least deprived of 2.40 (95% CI 1.36 to 4.27). In type 2 DM, the overall SIMD score was not significantly associated with increased prevalence of DR, with an OR for the most deprived relative to the least deprived of 0.85 (95% CI 0.71 to 1.02, p=0.07). CONCLUSIONS Socioeconomic deprivation is associated with increased prevalence of DR in patients with type 1 DM and this occurs earlier. This highlights the need for targeted interventions to address inequalities in eye healthcare.
Collapse
Affiliation(s)
- Liying Low
- Academic Unit of Ophthalmology, University of Birmingham, Birmingham and Midland Eye Centre, Birmingham, UK
| | | | - James Hodson
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Ritchie McAlpine
- Tayside Diabetes Managed Clinical Network (MCN), Ninewells Hospital and Medical School, Dundee, UK
| | - Una O'Colmain
- Department of Ophthalmology, Ninewells Hospital and Medical School, Dundee, UK
| | - Caroline MacEwen
- Department of Ophthalmology, Ninewells Hospital and Medical School, Dundee, UK
| |
Collapse
|
1569
|
Simmons KM, Michels AW. Type 1 diabetes: A predictable disease. World J Diabetes 2015; 6:380-390. [PMID: 25897349 PMCID: PMC4398895 DOI: 10.4239/wjd.v6.i3.380] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 11/26/2014] [Accepted: 01/12/2015] [Indexed: 02/05/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by loss of insulin producing beta cells and reliance on exogenous insulin for survival. T1D is one of the most common chronic diseases in childhood and the incidence is increasing, especially in children less than 5 years of age. In individuals with a genetic predisposition, an unidentified trigger initiates an abnormal immune response and the development of islet autoantibodies directed against proteins in insulin producing beta cells. There are currently four biochemical islet autoantibodies measured in the serum directed against insulin, glutamic decarboxylase, islet antigen 2, and zinc transporter 8. Development of islet autoantibodies occurs before clinical diagnosis of T1D, making T1D a predictable disease in an individual with 2 or more autoantibodies. Screening for islet autoantibodies is still predominantly done through research studies, but efforts are underway to screen the general population. The benefits of screening for islet autoantibodies include decreasing the incidence of diabetic ketoacidosis that can be life threatening, initiating insulin therapy sooner in the disease process, and evaluating safe and specific therapies in large randomized clinical intervention trials to delay or prevent progression to diabetes onset.
Collapse
|
1570
|
CCL20 is elevated during obesity and differentially regulated by NF-κB subunits in pancreatic β-cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2015; 1849:637-52. [PMID: 25882704 DOI: 10.1016/j.bbagrm.2015.03.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 03/05/2015] [Accepted: 03/30/2015] [Indexed: 12/20/2022]
Abstract
Enhanced leukocytic infiltration into pancreatic islets contributes to inflammation-based diminutions in functional β-cell mass. Insulitis (aka islet inflammation), which can be present in both T1DM and T2DM, is one factor influencing pancreatic β-cell death and dysfunction. IL-1β, an inflammatory mediator in both T1DM and T2DM, acutely (within 1h) induced expression of the CCL20 gene in rat and human islets and clonal β-cell lines. Transcriptional induction of CCL20 required the p65 subunit of NF-κB to replace the p50 subunit at two functional κB sites within the CCL20 proximal gene promoter. The NF-κB p50 subunit prevents CCL20 gene expression during unstimulated conditions and overexpression of p50 reduces CCL20, but enhances cyclooxygenase-2 (COX-2), transcript accumulation after exposure to IL-1β. We also identified differential recruitment of specific co-activator molecules to the CCL20 gene promoter, when compared with the CCL2 and COX2 genes, revealing distinct transcriptional requirements for individual NF-κB responsive genes. Moreover, IL-1β, TNF-α and IFN-γ individually increased the expression of CCR6, the receptor for CCL20, on the surface of human neutrophils. We further found that the chemokine CCL20 is elevated in serum from both genetically obese db/db mice and in C57BL6/J mice fed a high-fat diet. Taken together, these results are consistent with a possible activation of the CCL20-CCR6 axis in diseases with inflammatory components. Thus, interfering with this signaling pathway, either at the level of NF-κB-mediated chemokine production, or downstream receptor activation, could be a potential therapeutic target to offset inflammation-associated tissue dysfunction in obesity and diabetes.
Collapse
|
1571
|
Abstract
PURPOSE OF REVIEW The purpose of this article is to summarize recent developments in the histopathology of recent-onset type 1 diabetes. RECENT FINDINGS Insulitis is considered to be the defining lesion in young type 1 diabetic patients, and insight into its pathogenic mechanism is crucial for the development of effective new therapies. The present overview highlights some recent developments including an international consensus guideline for the diagnosis of insulitis in type 1 diabetic patients, immunophenotyping of the insulitic lesions, evidence for a heterogeneity of the disease process and a discussion on the differences and similarities between insulitis in patients and in rodent models of the disease. SUMMARY We have reviewed recent data, published over the last year, on the nature and mechanism of insulitis in both patients and the nonobese diabetic mouse model.
Collapse
Affiliation(s)
- Peter in't Veld
- Department of Pathology, Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
1572
|
Abstract
In the nearly 100 years since the discovery of therapeutic insulin, significant research efforts have been directed at finding the underlying cause of type 1 diabetes (T1D) and developing a "cure" for the disease. While progress has clearly been made toward each of these goals, neither vision has been fulfilled. With increasing pressure from both public and private funders of diabetes research, growing impatience of those with T1D at the lack of practical discoveries, increased competition for research funds, uncertainties on the reproducibility of published scientific data, and questions regarding the value of animal models, the current research environment has become extraordinarily difficult to traverse from the perspective of investigators. As a result, there is an increasing pressure toward performance of what might be considered "safe" research, where the aim is to affirm existing dogmas rather than to pioneer efforts involving unconventional thought. Psychologists refer to this practice as "observational bias" while cartoonists label the process the "streetlight effect." In this Perspective, we consider notions in T1D research that should be subject to bold question and provide additional concepts, many somewhat orphan to research efforts, whose investigation could lead to a means for truly identifying the cause of and a cure for T1D.
Collapse
Affiliation(s)
- Manuela Battaglia
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mark A Atkinson
- Departments of Pathology and Pediatrics, University of Florida, Gainesville, FL
| |
Collapse
|
1573
|
Abstract
Over the last years, there has been an increasing interest in the potential association between type 1 diabetes (T1D) and epilepsy. Both T1D and epilepsy are common conditions in children and adolescents, and therefore, their association might represent simply a coincidence or be related to common underlying mechanisms with a potential causal relationship. Few epidemiological studies have been performed in the pediatric population, and they have reached discordant conclusions, with some studies reporting an increased prevalence of epilepsy in children and adolescents with T1D, whereas others have not confirmed this finding. Several mechanisms could explain the occurrence of epilepsy in young people with T1D, such as metabolic abnormalities (hypo/hyperglycemia) and autoantibodies, along with a genetic predisposition and the presence of brain lesions/damage. Further studies are required to better define whether there is a causal relationship between the two conditions and to understand the underlying mechanisms.
Collapse
|
1574
|
Wang P, Fiaschi-Taesch NM, Vasavada RC, Scott DK, García-Ocaña A, Stewart AF. Diabetes mellitus--advances and challenges in human β-cell proliferation. Nat Rev Endocrinol 2015; 11:201-12. [PMID: 25687999 DOI: 10.1038/nrendo.2015.9] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The treatment of diabetes mellitus represents one of the greatest medical challenges of our era. Diabetes results from a deficiency or functional impairment of insulin-producing β cells, alone or in combination with insulin resistance. It logically follows that the replacement or regeneration of β cells should reverse the progression of diabetes and, indeed, this seems to be the case in humans and rodents. This concept has prompted attempts in many laboratories to create new human β cells using stem-cell strategies to transdifferentiate or reprogramme non-β cells into β cells or to discover small molecules or other compounds that can induce proliferation of human β cells. This latter approach has shown promise, but has also proven particularly challenging to implement. In this Review, we discuss the physiology of normal human β-cell replication, the molecular mechanisms that regulate the cell cycle in human β cells, the upstream intracellular signalling pathways that connect them to cell surface receptors on β cells, the epigenetic mechanisms that control human β-cell proliferation and unbiased approaches for discovering novel molecules that can drive human β-cell proliferation. Finally, we discuss the potential and challenges of implementing strategies that replace or regenerate β cells.
Collapse
Affiliation(s)
- Peng Wang
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Nathalie M Fiaschi-Taesch
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Rupangi C Vasavada
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Donald K Scott
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Adolfo García-Ocaña
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Andrew F Stewart
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, Atran 5, Box 1152, 1 Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
1575
|
Diana J, Lehuen A. Macrophages and β-cells are responsible for CXCR2-mediated neutrophil infiltration of the pancreas during autoimmune diabetes. EMBO Mol Med 2015; 6:1090-104. [PMID: 24968718 PMCID: PMC4154135 DOI: 10.15252/emmm.201404144] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Autoimmune type 1 diabetes (T1D) development results from the interaction between pancreatic β-cells, and the innate and the adaptive immune systems culminating with the destruction of the insulin-secreting β-cells by autoreactive T cells. This diabetogenic course starts during the first postnatal weeks by the infiltration of the pancreatic islets by innate immune cells and particularly neutrophils. Here, we aim to determine the cellular and molecular mechanism leading to the recruitment of this neutrophils in the pancreatic islets of non-obese diabetic (NOD) mice. Here, we show that neutrophil recruitment in the pancreatic islets is controlled by inflammatory macrophages and β-cells themselves. Macrophages and β-cells produce the chemokines CXCL1 and CXCL2, recruiting CXCR2-expressing neutrophils from the blood to the pancreatic islets. We further show that pancreatic macrophages secrete IL-1β-inducing CXCR2 ligand production by the β-cells. Finally, the blockade of neutrophil recruitment at early ages using CXCR2 antagonist dampens the diabetogenic T-cell response and the later development of autoimmune diabetes, supporting the therapeutic potential of this approach. Subject Categories Immunology; Metabolism
Collapse
Affiliation(s)
- Julien Diana
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, Necker-Enfants Malades Institute (INEM) Necker Hospital, Paris, France Sorbonne Paris Cité, Université Paris Descartes, Paris, France
| | - Agnès Lehuen
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France Institut National de la Santé et de la Recherche Médicale (INSERM), U1016, Cochin Institute Cochin Hospital, Paris, France Laboratoire d'Excellence INFLAMEX, Paris, France
| |
Collapse
|
1576
|
Garciafigueroa Y, Trucco M, Giannoukakis N. A brief glimpse over the horizon for type 1 diabetes nanotherapeutics. Clin Immunol 2015; 160:36-45. [PMID: 25817545 DOI: 10.1016/j.clim.2015.03.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 03/16/2015] [Indexed: 12/11/2022]
Abstract
The pace at which nanotherapeutic technology for human disease is evolving has accelerated exponentially over the past five years. Most of the technology is centered on drug delivery which, in some instances, offers tunable control of drug release. Emerging technologies have resulted in improvements in tissue and cell targeting while others are at the initial stages of pairing drug release and drug release kinetics with microenvironmental stimuli or changes in homeostasis. Nanotherapeutics has only recently been adopted for consideration as a prophylaxis/treatment approach in autoimmunity. Herein, we summarize the current state-of-the art of nanotherapeutics specifically for type 1 diabetes mellitus and offer our view over the horizon of where we envisage this modality evolving towards.
Collapse
Affiliation(s)
- Yesica Garciafigueroa
- Institute of Cellular Therapeutics, 11th Floor South Tower, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA 15212, USA.
| | - Massimo Trucco
- Institute of Cellular Therapeutics, 11th Floor South Tower, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA 15212, USA.
| | - Nick Giannoukakis
- Institute of Cellular Therapeutics, 11th Floor South Tower, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA 15212, USA.
| |
Collapse
|
1577
|
Regulatory vs. inflammatory cytokine T-cell responses to mutated insulin peptides in healthy and type 1 diabetic subjects. Proc Natl Acad Sci U S A 2015; 112:4429-34. [PMID: 25831495 DOI: 10.1073/pnas.1502967112] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Certain class II MHC (MHCII) alleles in mice and humans confer risk for or protection from type 1 diabetes (T1D). Insulin is a major autoantigen in T1D, but how its peptides are presented to CD4 T cells by MHCII risk alleles has been controversial. In the mouse model of T1D, CD4 T cells respond to insulin B-chain peptide (B:9-23) mimotopes engineered to bind the mouse MHCII molecule, IA(g7), in an unfavorable position or register. Because of the similarities between IA(g7) and human HLA-DQ T1D risk alleles, we examined control and T1D subjects with these risk alleles for CD4 T-cell responses to the same natural B:9-23 peptide and mimotopes. A high proportion of new-onset T1D subjects mounted an inflammatory IFN-γ response much more frequently to one of the mimotope peptides than to the natural peptide. Surprisingly, the control subjects bearing an HLA-DQ risk allele also did. However, these control subjects, especially those with only one HLA-DQ risk allele, very frequently made an IL-10 response, a cytokine associated with regulatory T cells. T1D subjects with established disease also responded to the mimotope rather than the natural B:9-23 peptide in proliferation assays and the proliferating cells were highly enriched in certain T-cell receptor sequences. Our results suggest that the risk of T1D may be related to how an HLA-DQ genotype determines the balance of T-cell inflammatory vs. regulatory responses to insulin, having important implications for the use and monitoring of insulin-specific therapies to prevent diabetes onset.
Collapse
|
1578
|
Hayes CE, Hubler SL, Moore JR, Barta LE, Praska CE, Nashold FE. Vitamin D Actions on CD4(+) T Cells in Autoimmune Disease. Front Immunol 2015; 6:100. [PMID: 25852682 PMCID: PMC4364365 DOI: 10.3389/fimmu.2015.00100] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/23/2015] [Indexed: 12/11/2022] Open
Abstract
This review summarizes and integrates research on vitamin D and CD4+ T-lymphocyte biology to develop new mechanistic insights into the molecular etiology of autoimmune disease. A deep understanding of molecular mechanisms relevant to gene–environment interactions is needed to deliver etiology-based autoimmune disease prevention and treatment strategies. Evidence linking sunlight, vitamin D, and the risk of multiple sclerosis and type 1 diabetes is summarized to develop the thesis that vitamin D is the environmental factor that most strongly influences autoimmune disease development. Evidence for CD4+ T-cell involvement in autoimmune disease pathogenesis and for paracrine calcitriol signaling to CD4+ T lymphocytes is summarized to support the thesis that calcitriol is sunlight’s main protective signal transducer in autoimmune disease risk. Animal modeling and human mechanistic data are summarized to support the view that vitamin D probably influences thymic negative selection, effector Th1 and Th17 pathogenesis and responsiveness to extrinsic cell death signals, FoxP3+CD4+ T-regulatory cell and CD4+ T-regulatory cell type 1 (Tr1) cell functions, and a Th1–Tr1 switch. The proposed Th1–Tr1 switch appears to bridge two stable, self-reinforcing immune states, pro- and anti-inflammatory, each with a characteristic gene regulatory network. The bi-stable switch would enable T cells to integrate signals from pathogens, hormones, cell–cell interactions, and soluble mediators and respond in a biologically appropriate manner. Finally, unanswered questions and potentially informative future research directions are highlighted to speed delivery of etiology-based strategies to reduce autoimmune disease.
Collapse
Affiliation(s)
- Colleen Elizabeth Hayes
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison , Madison, WI , USA
| | - Shane L Hubler
- Department of Statistics, College of Letters and Sciences, University of Wisconsin-Madison , Madison, WI , USA
| | - Jerott R Moore
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison , Madison, WI , USA
| | - Lauren E Barta
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison , Madison, WI , USA
| | - Corinne E Praska
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison , Madison, WI , USA
| | - Faye E Nashold
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison , Madison, WI , USA
| |
Collapse
|
1579
|
Aires MB, Santos ACVD. Effects of maternal diabetes on trophoblast cells. World J Diabetes 2015; 6:338-344. [PMID: 25789116 PMCID: PMC4360428 DOI: 10.4239/wjd.v6.i2.338] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 11/15/2014] [Accepted: 12/31/2014] [Indexed: 02/05/2023] Open
Abstract
Diabetes mellitus (DM) is a health condition characterized by hyperglycemia over a prolonged period. There are three main types of DM: DM type 1 (DM1), DM2 and gestational DM (GDM). Maternal diabetes, which includes the occurrence of DM1 and DM2 during pregnancy or GDM, increases the occurrence of gesttional complications and adverse fetal outcomes. The hyperglycemic intrauterine environment affects not only the fetus but also the placental development and function in humans and experimental rodents. The underlying mechanisms are still unclear, but some evidence indicates alterations in trophoblast proliferation, apoptosis and cell cycle control in diabetes. A proper coordination of trophoblast proliferation, differentiation and invasion is required for placental development. Initially, increased expression of proliferative markers in junctional and labyrinth zones of rat placentas and villous cytotrophoblast, syncytiotrophoblast, stromal cells and fetal endothelial cells in human placentas is reported among diabetics. Moreover, reduced apoptotic index and expression of some apoptotic genes are described in placentas of GDM women. In addition, cell cycle regulators including cyclins and cyclin-dependent kinase inhibitors seem to be affected by the hyperglycemic environment. More studies are necessary to check the balance between proliferation, apoptosis and differentiation in trophoblast cells during maternal diabetes.
Collapse
|
1580
|
Engman C, Wen Y, Meng WS, Bottino R, Trucco M, Giannoukakis N. Generation of antigen-specific Foxp3+ regulatory T-cells in vivo following administration of diabetes-reversing tolerogenic microspheres does not require provision of antigen in the formulation. Clin Immunol 2015; 160:103-23. [PMID: 25773782 DOI: 10.1016/j.clim.2015.03.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 03/05/2015] [Indexed: 11/20/2022]
Abstract
We have developed novel antisense oligonucleotide-formulated microspheres that can reverse hyperglycemia in newly-onset diabetic mice. Dendritic cells taking up the microspheres adopt a restrained co-stimulation ability and migrate to the pancreatic lymph nodes when injected into an abdominal region that is drained by those lymph nodes. Furthermore, we demonstrate that the absolute numbers of antigen-specific Foxp3+ T regulatory cells are increased only in the lymph nodes draining the site of administration and that these T-cells proliferate independently of antigen supply in the microspheres. Taken together, our data add to the emerging model where antigen supply may not be a requirement in "vaccines" for autoimmune disease, but the site of administration - subserved by lymph nodes draining the target organ - is in fact critical to foster the generation of antigen-specific regulatory cells. The implications of these observations on "vaccine" design for autoimmunity are discussed and summarized.
Collapse
MESH Headings
- Animals
- B7-1 Antigen/genetics
- B7-2 Antigen/genetics
- Blood Glucose/drug effects
- CD11c Antigen/metabolism
- CD40 Antigens/genetics
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Dendritic Cells/immunology
- Diabetes Mellitus, Experimental/therapy
- Diabetes Mellitus, Type 1/therapy
- Female
- Forkhead Transcription Factors/analysis
- Gene Knockdown Techniques
- Hyperglycemia/therapy
- Leukocyte Common Antigens/metabolism
- Lymph Nodes/cytology
- Lymph Nodes/immunology
- Lymphocyte Activation/immunology
- Macaca fascicularis
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred NOD
- Mice, Transgenic
- Microspheres
- Oligonucleotides, Antisense/genetics
- Pancreas/immunology
- Receptors, Antigen, T-Cell/genetics
- T-Lymphocytes, Regulatory/cytology
- Vaccines/administration & dosage
- Vaccines/immunology
Collapse
Affiliation(s)
- Carl Engman
- Institute of Cellular Therapeutics, 11th Floor South Tower, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA, 15212, USA.
| | - Yi Wen
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Mellon 413, 600 Forbes Avenue, Pittsburgh, PA 15282, USA
| | - Wilson S Meng
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Mellon 413, 600 Forbes Avenue, Pittsburgh, PA 15282, USA.
| | - Rita Bottino
- Institute of Cellular Therapeutics, 11th Floor South Tower, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA, 15212, USA.
| | - Massimo Trucco
- Institute of Cellular Therapeutics, 11th Floor South Tower, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA, 15212, USA.
| | - Nick Giannoukakis
- Institute of Cellular Therapeutics, 11th Floor South Tower, Allegheny Health Network, 320 East North Avenue, Pittsburgh, PA, 15212, USA.
| |
Collapse
|
1581
|
Heywood J, Evangelou M, Goymer D, Kennet J, Anselmiova K, Guy C, O'Brien C, Nutland S, Brown J, Walker NM, Todd JA, Waldron-Lynch F. Effective recruitment of participants to a phase I study using the internet and publicity releases through charities and patient organisations: analysis of the adaptive study of IL-2 dose on regulatory T cells in type 1 diabetes (DILT1D). Trials 2015; 16:86. [PMID: 25881192 PMCID: PMC4369347 DOI: 10.1186/s13063-015-0583-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/27/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND A barrier to the successful development of new disease treatments is the timely recruitment of participants to experimental medicine studies that are primarily designed to investigate biological mechanisms rather than evaluate clinical efficacy. The aim of this study was to analyse the performance of three recruitment sources and the effect of publicity events during the Adaptive study of IL-2 dose on regulatory T cells in type 1 diabetes (DILT1D). METHODS The final study outcome, demography, disease duration, residence and the effect of publicity events on the performance of three recruitment sources (clinics, type 1 diabetes (T1D) disease register and the internet) were analysed from a bespoke DILT1D recruitment database. For the internet source, the origin of website hits in relation to publicity events was also evaluated. RESULTS A total of 735 potentially eligible participants were approached to identify the final 45 DILT1D participants. A total of 477 (64%) were identified via the disease register, but only 59 (12%) responded to contact. A total of 317 individuals registered with the DILT1D study team. Self-referral via the study website generated 170 (54%) registered individuals and was the most popular and successful source, with 88 (28%) sourced from diabetes clinics and 59 (19%) from the disease register. Of those with known T1D duration (N = 272), the internet and clinics sources identified a larger number (57, 21%) of newly diagnosed T1D (<100 days post-diagnosis) compared to the register (1, 0.4%). The internet extended the geographical reach of the study, enabling both national and international participation. Targeted website posts and promotional events from organisations supporting T1D research and treatment during the trial were essential to the success of the internet recruitment strategy. CONCLUSIONS Analysis of the DILT1D study recruitment outcomes illustrates the utility of an active internet recruitment strategy, supported by patient groups and charities, funding agencies and sponsors, in successfully conducting an early phase study in T1D. This recruitment strategy should now be evaluated in late-stage trials to develop treatments for T1D and other diseases. TRIAL REGISTRATION NCT01827735 (registered: 4 April 2013); ISRCTN27852285 (registered: 23 March 2013); DRN767 (registered: 21 January 2013).
Collapse
Affiliation(s)
- James Heywood
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
| | - Marina Evangelou
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
| | - Donna Goymer
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
| | - Jane Kennet
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
| | - Katerina Anselmiova
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
| | - Catherine Guy
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
| | - Criona O'Brien
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
| | - Sarah Nutland
- The Cambridge BioResource, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.
| | - Judy Brown
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
| | - Neil M Walker
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
| | - John A Todd
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
| | - Frank Waldron-Lynch
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, NIHR Cambridge Biomedical Research Centre, Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
| |
Collapse
|
1582
|
Abstract
Reactive oxygen species (ROS) can cause pancreatic β-cell death by activating transient receptor potential (melastatin) 2 (TRPM2) channels. Cell death has been attributed to the ability of these channels to raise cytosolic Ca2+. Recent studies however revealed that TRPM2 channels can also conduct Zn2+, but the physiological relevance of this property is enigmatic. Given that Zn2+ is cytotoxic, we asked whether TRPM2 channels can permeate sufficient Zn2+ to affect cell viability. To address this, we used the insulin secreting (INS1) β-cell line, human embryonic kidney (HEK)-293 cells transfected with TRPM2 and pancreatic islets. H2O2 activation of TRPM2 channels increases the cytosolic levels of both Ca2+ and Zn2+ and causes apoptotic cell death. Interestingly, chelation of Zn2+ alone was sufficient to prevent β-cell death. The source of the cytotoxic Zn2+ is intracellular, found largely sequestered in lysosomes. Lysosomes express TRPM2 channels, providing a potential route for Zn2+ release. Zn2+ release is potentiated by extracellular Ca2+ entry, indicating that Ca2+-induced Zn2+ release leads to apoptosis. Knockout of TRPM2 channels protects mice from β-cell death and hyperglycaemia induced by multiple low-dose streptozotocin (STZ; MLDS) administration. These results argue that TRPM2-mediated, Ca2+-potentiated Zn2+ release underlies ROS-induced β-cell death and Zn2+, rather than Ca2+, plays a primary role in apoptosis.
Collapse
|
1583
|
Taylor-Fishwick DA, Weaver J, Glenn L, Kuhn N, Rai G, Jadhav A, Simeonov A, Dudda A, Schmoll D, Holman TR, Maloney DJ, Nadler JL. Selective inhibition of 12-lipoxygenase protects islets and beta cells from inflammatory cytokine-mediated beta cell dysfunction. Diabetologia 2015; 58:549-57. [PMID: 25417214 DOI: 10.1007/s00125-014-3452-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 10/23/2014] [Indexed: 12/30/2022]
Abstract
AIMS/HYPOTHESIS Islet inflammation leads to loss of functional pancreatic beta cell mass. Increasing evidence suggests that activation of 12-lipoxygenase leads to inflammatory beta cell loss. This study evaluates new specific small-molecule inhibitors of 12-lipoxygenase for protecting rodent and human beta cells from inflammatory damage. METHODS Mouse beta cell lines and mouse and human islets were treated with inflammatory cytokines IL-1β, TNFα and IFNγ in the absence or presence of novel selective 12-lipoxygenase inhibitors. Glucose-stimulated insulin secretion (GSIS), gene expression, cell survival and 12-S-hydroxyeicosatetraenoic acid (12-S-HETE) levels were evaluated using established methods. Pharmacokinetic analysis was performed with the lead inhibitor in CD1 mice. RESULTS Inflammatory cytokines led to the loss of human beta cell function, elevated cell death, increased inflammatory gene expression and upregulation of 12-lipoxygenase expression and activity (measured by 12-S-HETE generation). Two 12-lipoxygenase inhibitors, Compounds 5 and 9, produced a concentration-dependent reduction of stimulated 12-S-HETE levels. GSIS was preserved in the presence of the 12-lipoxygenase inhibitors. 12-Lipoxygenase inhibition preserved survival of primary mouse and human islets. When administered orally, Compound 5 reduced plasma 12-S-HETE in CD1 mice. Compounds 5 and 9 preserved the function and survival of human donor islets exposed to inflammatory cytokines. CONCLUSIONS/INTERPRETATION Selective inhibition of 12-lipoxygenase activity confers protection to beta cells during exposure to inflammatory cytokines. These concept validation studies identify 12-lipoxygenase as a promising target in the prevention of loss of functional beta cells in diabetes.
Collapse
Affiliation(s)
- David A Taylor-Fishwick
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 W. Olney Road, Norfolk, VA, 23507, USA,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1584
|
Growth attainments of Indian children with type 1 diabetes: a mixed longitudinal study. Indian J Pediatr 2015; 82:245-52. [PMID: 24827083 DOI: 10.1007/s12098-014-1466-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 04/17/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To evaluate distance and velocity growth of children with Type 1 Diabetes (T1D) and quantify the magnitude of gender differences in anthropometric parameters. METHODS Children with T1D, without any coexisting condition that could affect growth were measured at half yearly intervals for body weight, height, occipito-frontal circumference (OFC), chest circumference (CC), mid upper arm circumference (MUAC), triceps skinfold thickness (TSFT), biceps skinfold thickness (BSFT) and subscapular skinfold thickness (SSSFT) using standardized techniques. Comparison of these growth parameters was made between the 2 genders as well as with the published norms. RESULTS Of the 115 children studied, there were no significant gender differences in weight at different age points. Boys measured taller than girls but the gender differences in height were significant only at 7 and 10 y. There was a tendency of higher BMIs in girls. In comparison to WHO standards and normal Indian children, boys had lower weight initially but became heavier later. The girls showed close similarity to their normal Indian counterparts but remained lighter than their WHO counterparts. After the initial lower height, an acceleratory trend was noted in boys but girls remained shorter throughout the study as compared to WHO peers. Boys, in general had higher CC, OFC, MUAC and MUAMC. The skinfold thicknesses recorded inconsistent growth patterns in both sexes. Growth velocities for various body parameters except skinfold thicknesses were similar in boys and girls. CONCLUSIONS Initial weight and height loss followed by catch up growth was noted in boys. The values of MUAC, MUAMC and skinfold thicknesses as compared to data from developed countries, were lower in present patients.
Collapse
|
1585
|
Hansen MP, Matheis N, Kahaly GJ. Type 1 diabetes and polyglandular autoimmune syndrome: A review. World J Diabetes 2015; 6:67-79. [PMID: 25685279 PMCID: PMC4317318 DOI: 10.4239/wjd.v6.i1.67] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 11/11/2014] [Accepted: 12/01/2014] [Indexed: 02/05/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disorder caused by inflammatory destruction of the pancreatic tissue. The etiopathogenesis and characteristics of the pathologic process of pancreatic destruction are well described. In addition, the putative susceptibility genes for T1D as a monoglandular disease and the relation to polyglandular autoimmune syndrome (PAS) have also been well explored. The incidence of T1D has steadily increased in most parts of the world, especially in industrialized nations. T1D is frequently associated with autoimmune endocrine and non-endocrine diseases and patients with T1D are at a higher risk for developing several glandular autoimmune diseases. Familial clustering is observed, which suggests that there is a genetic predisposition. Various hypotheses pertaining to viral- and bacterial-induced pancreatic autoimmunity have been proposed, however a definitive delineation of the autoimmune pathomechanism is still lacking. In patients with PAS, pancreatic and endocrine autoantigens either colocalize on one antigen-presenting cell or are expressed on two/various target cells sharing a common amino acid, which facilitates binding to and activation of T cells. The most prevalent PAS phenotype is the adult type 3 variant or PAS type III, which encompasses T1D and autoimmune thyroid disease. This review discusses the findings of recent studies showing noticeable differences in the genetic background and clinical phenotype of T1D either as an isolated autoimmune endocrinopathy or within the scope of polyglandular autoimmune syndrome.
Collapse
|
1586
|
Brozzi F, Gerlo S, Grieco FA, Nardelli TR, Lievens S, Gysemans C, Marselli L, Marchetti P, Mathieu C, Tavernier J, Eizirik DL. A combined "omics" approach identifies N-Myc interactor as a novel cytokine-induced regulator of IRE1 protein and c-Jun N-terminal kinase in pancreatic beta cells. J Biol Chem 2015; 289:20677-93. [PMID: 24936061 DOI: 10.1074/jbc.m114.568808] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Type 1 diabetes is an autoimmune disease with a strong inflammatory component. The cytokines interleukin-1β and interferon-γ contribute to beta cell apoptosis in type 1 diabetes. These cytokines induce endoplasmic reticulum stress and the unfolded protein response (UPR), contributing to the loss of beta cells. IRE1α, one of the UPR mediators, triggers insulin degradation and inflammation in beta cells and is critical for the transition from "physiological" to "pathological" UPR. The mechanisms regulating inositol-requiring protein 1α (IRE1α) activation and its signaling for beta cell "adaptation," "stress response," or "apoptosis" remain to be clarified. To address these questions, we combined mammalian protein-protein interaction trap-based IRE1α interactome and functional genomic analysis of human and rodent beta cells exposed to pro-inflammatory cytokines to identify novel cytokine-induced regulators of IRE1α. Based on this approach, we identified N-Myc interactor (NMI) as an IRE1α-interacting/modulator protein in rodent and human pancreatic beta cells. An increased expression of NMI was detected in islets from nonobese diabetic mice with insulitis and in rodent or human beta cells exposed in vitro to the pro-inflammatory cytokines interleukin-1β and interferon-γ. Detailed mechanistic studies demonstrated that NMI negatively modulates IRE1α-dependent activation of JNK and apoptosis in rodent and human pancreatic beta cells. In conclusion, by using a combined omics approach, we identified NMI induction as a novel negative feedback mechanism that decreases IRE1α-dependent activation of JNK and apoptosis in cytokine-exposed beta cells
Collapse
|
1587
|
Sacks DB, Kricka LJ. Plasmonic chip tackles type 1 diabetes diagnosis. Clin Chem 2015; 61:794-6. [PMID: 25655270 DOI: 10.1373/clinchem.2014.233346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 12/03/2014] [Indexed: 11/06/2022]
Affiliation(s)
- David B Sacks
- Department of Laboratory Medicine, NIH, Bethesda, MD
| | | |
Collapse
|
1588
|
Lund-Blix NA, Stene LC, Rasmussen T, Torjesen PA, Andersen LF, Rønningen KS. Infant feeding in relation to islet autoimmunity and type 1 diabetes in genetically susceptible children: the MIDIA Study. Diabetes Care 2015; 38:257-63. [PMID: 25422170 DOI: 10.2337/dc14-1130] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE We aimed to study the association of breast-feeding duration and age at the introduction of solid foods with the risk of islet autoimmunity and type 1 diabetes in genetically susceptible children. RESEARCH DESIGN AND METHODS Newborns were recruited from the Norwegian general population during 2001-2007. After genetic screening of nearly 50,000 newborns, 908 children with the high-risk HLA genotype were followed up with blood samples and questionnaires at age 3, 6, 9, and 12 months and then annually. Complete infant diet data were available for 726 children. RESULTS Any breast-feeding for 12 months or longer predicted a decreased risk of developing type 1 diabetes compared with any breast-feeding for less than 12 months before and after adjusting for having a first-degree relative with type 1 diabetes, vitamin D supplementation, maternal education, sex, and delivery type (hazard ratio 0.37 [95% CI 0.15-0.93]). Any breast-feeding for 12 months or longer was not associated with islet autoimmunity but predicted a lower risk of progression from islet autoimmunity to type 1 diabetes (hazard ratio 0.35 [95% CI 0.13-0.94]). Duration of full breast-feeding was not significantly associated with the risk of islet autoimmunity or type 1 diabetes nor was age at introduction of solid foods or breast-feeding at the time of introduction of any solid foods. CONCLUSIONS These results suggest that breast-feeding for 12 months or longer predict a lower risk of progression from islet autoimmunity to type 1 diabetes among genetically predisposed children.
Collapse
Affiliation(s)
- Nicolai A Lund-Blix
- Department of Pediatric Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway Department of Nutrition, University of Oslo, Oslo, Norway
| | - Lars C Stene
- Division of Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Trond Rasmussen
- Division of Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Peter A Torjesen
- Hormone Laboratory, Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | | | - Kjersti S Rønningen
- Department of Pediatric Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
1589
|
Woittiez NJC, Roep BO. Impact of disease heterogeneity on treatment efficacy of immunotherapy in Type 1 diabetes: different shades of gray. Immunotherapy 2015; 7:163-74. [DOI: 10.2217/imt.14.104] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes results from selective destruction of insulin-producing pancreatic β-cells by a progressive autoimmune process. Type 1 diabetes proves very heterogeneous in pathology, disease progression and efficacy of therapeutic intervention. Indeed, several immunotherapies that appear ineffective for the entire treated patient population in fact look promising in subgroups of patients. It therefore seems inconceivable that one standard therapy will provide the golden bullet of disease intervention. Instead, personalized medicine may improve immune intervention efficacy rates. We discuss the effect of disease heterogeneity on treatment outcome of immunotherapies, identifying apparent gaps in our understanding of treatment efficacy in subgroups of Type 1 diabetic patients as well as identifying future opportunities for immunotherapy.
Collapse
Affiliation(s)
- Nicky JC Woittiez
- Department of Immunohematology & Blood Transfusion, Leiden University Medical Center, E3-Q, LUMC, PO Box 9600, NL-2300RC Leiden, The Netherlands
| | - Bart O Roep
- Department of Immunohematology & Blood Transfusion, Leiden University Medical Center, E3-Q, LUMC, PO Box 9600, NL-2300RC Leiden, The Netherlands
| |
Collapse
|
1590
|
Kumar RB, Gupta M, Feldman BJ. The development of next-generation screening and diagnostic platforms will change diabetes care. Expert Rev Mol Diagn 2015; 15:291-4. [PMID: 25583407 DOI: 10.1586/14737159.2015.1002468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Diabetes mellitus is a common disease with a rising incidence and the findings of hyperglycemia and glucosuria. However, there are multiple types of diabetes, each with distinct etiologies. The two major types of diabetes are type 1, which is caused by an autoimmune process, and type 2, which is thought to be primarily metabolic, resulting from insulin resistance, often in the setting of obesity. Historically, the distinction between these two types was obvious. Here, we discuss how this paradigm has dramatically changed because of both the evolving epidemiology of diabetes mellitus and new and emerging tools, and therapies to diagnose and treat diabetes. As we believe that understanding these changes is critical to providing optimal care to patients with diabetes, we have developed a novel plasmonic gold chip platform that is able to meet the new and emerging demands of modern diabetes care.
Collapse
Affiliation(s)
- Rajiv B Kumar
- Department of Pediatrics, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | | | | |
Collapse
|
1591
|
Wang Q, Guo T, Portas J, McPherron AC. A soluble activin receptor type IIB does not improve blood glucose in streptozotocin-treated mice. Int J Biol Sci 2015; 11:199-208. [PMID: 25561902 PMCID: PMC4279095 DOI: 10.7150/ijbs.10430] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 11/05/2014] [Indexed: 12/18/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM), or insulin dependent DM, is accompanied by decreased muscle mass. The growth factor myostatin (MSTN) is a negative regulator of muscle growth, and a loss of MSTN signaling has been shown to increase muscle mass and prevent the development of obesity, insulin resistance and lipodystrophic diabetes in mice. The effects of MSTN inhibition in a T1DM model on muscle mass and blood glucose are unknown. We asked whether MSTN inhibition would increase muscle mass and decrease hyperglycemia in mice treated with streptozotocin (STZ) to destroy pancreatic beta cells. After diabetes developed, mice were treated with a soluble MSTN/activin receptor fused to Fc (ACVR2B:Fc). ACVR2B:Fc increased body weight and muscle mass compared to vehicle treated mice. Unexpectedly, ACVR2B:Fc reproducibly exacerbated hyperglycemia within approximately one week of administration. ACVR2B:Fc treatment also elevated serum levels of the glucocorticoid corticosterone. These results suggest that although MSTN/activin inhibitors increased muscle mass, they may be counterproductive in improving health in patients with T1DM.
Collapse
Affiliation(s)
- Qian Wang
- 1. Current Addresses: Pathology Department, Stony Brook University Medical Center, Stony Brook, New York, USA
| | - Tingqing Guo
- 2. Novo Nordisk Research Centre China, Changping District, Beijing, China
| | - Jennifer Portas
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
1592
|
Powell DR, Doree D, Jeter-Jones S, Ding ZM, Zambrowicz B, Sands A. Sotagliflozin improves glycemic control in nonobese diabetes-prone mice with type 1 diabetes. Diabetes Metab Syndr Obes 2015; 8:121-7. [PMID: 25759591 PMCID: PMC4346285 DOI: 10.2147/dmso.s76342] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
PURPOSE Oral agents are needed that improve glycemic control without increasing hypoglycemic events in patients with type 1 diabetes (T1D). Sotagliflozin may meet this need, because this compound lowers blood glucose through the insulin-independent mechanisms of inhibiting kidney SGLT2 and intestinal SGLT1. We examined the effect of sotagliflozin on glycemic control and rate of hypoglycemia measurements in T1D mice maintained on a low daily insulin dose, and compared these results to those from mice maintained in better glycemic control with a higher daily insulin dose alone. MATERIALS AND METHODS Nonobese diabetes-prone mice with cyclophosphamide-induced T1D were randomized to receive one of four daily treatments: 0.2 U insulin/vehicle, 0.05 U insulin/vehicle, 0.05 U insulin/2 mg/kg sotagliflozin or 0.05 U insulin/30 mg/kg sotagliflozin. Insulin was delivered subcutaneously by micro-osmotic pump; the day after pump implantation, mice received their first of 22 once-daily oral doses of sotagliflozin or vehicle. Glycemic control was monitored by measuring fed blood glucose and hemoglobin A1c levels. RESULTS Blood glucose levels decreased rapidly and comparably in the 0.05 U insulin/sotagliflozin-treated groups and the 0.2 U insulin/vehicle group compared to the 0.05 U insulin/vehicle group, which had significantly higher levels than the other three groups from day 2 through day 23. A1c levels were also significantly higher in the 0.05 U insulin/vehicle group compared to the other three groups on day 23. Importantly, the 0.2 U insulin/vehicle group had, out of 100 blood glucose measurements, 13 that were <70 mg/dL compared to one of 290 for the other three groups combined. CONCLUSION Sotagliflozin significantly improved glycemic control, without increasing the rate of hypoglycemia measurements, in diabetic mice maintained on a low insulin dose. This sotagliflozin-mediated improvement in glycemic control was comparable to that achieved by raising the insulin dose alone, but was not accompanied by the increased rate of hypoglycemia measurements observed with the higher insulin dose.
Collapse
Affiliation(s)
- David R Powell
- Lexicon Pharmaceuticals, The Woodlands, TX, USA
- Correspondence: David R Powell, Lexicon Pharmaceuticals, 8800 Technology Forest Place, The Woodlands, TX 77381-1160, USA, Tel +1 281 863 3060, Fax +1 281 863 8115, Email
| | - Deon Doree
- Lexicon Pharmaceuticals, The Woodlands, TX, USA
| | | | | | | | | |
Collapse
|
1593
|
Hilliard ME, Anderson BJ. Contemporary faces of diabetes care for youth and young adults in the 21st century: evolution in the roles of patients and families, healthcare providers and systems, behavioral health, and the online community. Curr Diabetes Rev 2015; 11:208-9. [PMID: 26239946 PMCID: PMC4582404 DOI: 10.2174/1573399811999150706115014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Marisa E. Hilliard
- Current Diabeties Reviewes, Baylor College of Medicine, Department of Pediatrics, Houston, Texas, USA, 77030,
| | - Barbara J. Anderson
- Current Diabeties Reviewes, Baylor College of Medicine, Department of Pediatrics, Houston, Texas, USA, 77030,
| |
Collapse
|
1594
|
Abstract
This article highlights the difficulties in creating a definitive classification of diabetes mellitus in the absence of a complete understanding of the pathogenesis of the major forms. This brief review shows the evolving nature of the classification of diabetes mellitus. No classification scheme is ideal, and all have some overlap and inconsistencies. The only diabetes in which it is possible to accurately diagnose by DNA sequencing, monogenic diabetes, remains undiagnosed in more than 90% of the individuals who have diabetes caused by one of the known gene mutations. The point of classification, or taxonomy, of disease, should be to give insight into both pathogenesis and treatment. It remains a source of frustration that all schemes of diabetes mellitus continue to fall short of this goal.
Collapse
Affiliation(s)
- Celeste C Thomas
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, The University of Chicago, 5841 South Maryland Avenue, MC 1027, Chicago, IL 60637, USA.
| | - Louis H Philipson
- Department of Medicine, Section of Endocrinology, Diabetes and Metabolism, The University of Chicago, 5841 South Maryland Avenue, MC 1027, Chicago, IL 60637, USA; Department of Pediatrics, Section of Endocrinology, Diabetes and Metabolism, The University of Chicago, 900 East 57th Street, Chicago, IL 60637, USA
| |
Collapse
|
1595
|
Ilonen J, Knip M, Vaarala O. Heterogeneity in diabetes-associated autoantibodies and susceptibility to Type 1 diabetes: lessons for disease prevention. Expert Rev Endocrinol Metab 2015; 10:25-34. [PMID: 30289041 DOI: 10.1586/17446651.2015.955474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Autoantibodies against pancreatic islets are strong predictors of Type 1 diabetes. When persistent β-cell autoantibodies against at least two autoantigens are detected, the probability of diabetes is extremely high, although the time period before disease development can vary from days up to more than 20 years. Insulin autoantibodies or antibodies specific to glutamate decarboxylase 65 enzyme are in most cases, the first autoantibodies to appear. Insulin autoantibodies typically emerge very early with a peak at the age of 1.5 years, whereas the onset of glutamic acid decarboxylase 65 antibody positivity has a more even distribution, peaking later in childhood. These differences in the timing of appearance suggest that different environmental factors might be involved in the initiation of β-cell autoimmunity beginning either already in infancy or later on. This should be taken into account in studies aimed at identifying environmental factors triggering islet cell-specific autoimmunity and also in the design of prevention trials.
Collapse
Affiliation(s)
- Jorma Ilonen
- a 1 Immunogenetics Laboratory, University of Turku, Turku, Finland
- b 2 Department of Clinical Microbiology, University of Eastern Finland, Kuopio, Finland
| | - Mikael Knip
- c 3 Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- d 4 Diabetes and Obesity Research Program, University of Helsinki, Helsinki, Finland
- e 5 Folkhälsan Research Center, Helsinki, Finland
- f 6 Department of Pediatrics, Tampere University Hospital, Tampere, Finland
| | - Outi Vaarala
- g 7 Department of Vaccination and Immune Protection, National Institute for Health and Welfare, Helsinki, Finland
| |
Collapse
|
1596
|
Haller MJ, Gitelman SE, Gottlieb PA, Michels AW, Rosenthal SM, Shuster JJ, Zou B, Brusko TM, Hulme MA, Wasserfall CH, Mathews CE, Atkinson MA, Schatz DA. Anti-thymocyte globulin/G-CSF treatment preserves β cell function in patients with established type 1 diabetes. J Clin Invest 2014; 125:448-55. [PMID: 25500887 DOI: 10.1172/jci78492] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/31/2014] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Previous efforts to preserve β cell function in individuals with type 1 diabetes (T1D) have focused largely on the use of single immunomodulatory agents administered within 100 days of diagnosis. Based on human and preclinical studies, we hypothesized that a combination of low-dose anti-thymocyte globulin (ATG) and pegylated granulocyte CSF (G-CSF) would preserve β cell function in patients with established T1D (duration of T1D >4 months and <2 years). METHODS A randomized, single-blinded, placebo-controlled trial was performed on 25 subjects: 17 subjects received ATG (2.5 mg/kg intravenously) followed by pegylated G-CSF (6 mg subcutaneously every 2 weeks for 6 doses) and 8 subjects received placebo. The primary outcome was the 1-year change in AUC C-peptide following a 2-hour mixed-meal tolerance test (MMTT). At baseline, the age (mean ± SD) was 24.6 ± 10 years; mean BMI was 25.4 ± 5.2 kg/m²; mean A1c was 6.5% ± 1.1%; insulin use was 0.31 ± 0.22 units/kg/d; and length of diagnosis was 1 ± 0.5 years. RESULTS Combination ATG/G-CSF treatment tended to preserve β cell function in patients with established T1D. The mean difference in MMTT-stimulated AUC C-peptide between treated and placebo subjects was 0.28 nmol/l/min (95% CI 0.001-0.552, P = 0.050). A1c was lower in ATG/G-CSF-treated subjects at the 6-month study visit. ATG/G-CSF therapy was associated with relative preservation of Tregs. CONCLUSIONS Patients with established T1D may benefit from combination immunotherapy approaches to preserve β cell function. Further studies are needed to determine whether such approaches may prevent or delay the onset of the disease. TRIAL REGISTRATION Clinicaltrials.gov NCT01106157. FUNDING The Leona M. and Harry B. Helmsley Charitable Trust and Sanofi.
Collapse
|
1597
|
Davis-Richardson AG, Ardissone AN, Dias R, Simell V, Leonard MT, Kemppainen KM, Drew JC, Schatz D, Atkinson MA, Kolaczkowski B, Ilonen J, Knip M, Toppari J, Nurminen N, Hyöty H, Veijola R, Simell T, Mykkänen J, Simell O, Triplett EW. Bacteroides dorei dominates gut microbiome prior to autoimmunity in Finnish children at high risk for type 1 diabetes. Front Microbiol 2014; 5:678. [PMID: 25540641 PMCID: PMC4261809 DOI: 10.3389/fmicb.2014.00678] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/19/2014] [Indexed: 01/15/2023] Open
Abstract
The incidence of the autoimmune disease, type 1 diabetes (T1D), has increased dramatically over the last half century in many developed countries and is particularly high in Finland and other Nordic countries. Along with genetic predisposition, environmental factors are thought to play a critical role in this increase. As with other autoimmune diseases, the gut microbiome is thought to play a potential role in controlling progression to T1D in children with high genetic risk, but we know little about how the gut microbiome develops in children with high genetic risk for T1D. In this study, the early development of the gut microbiomes of 76 children at high genetic risk for T1D was determined using high-throughput 16S rRNA gene sequencing. Stool samples from children born in the same hospital in Turku, Finland were collected at monthly intervals beginning at 4-6 months after birth until 2.2 years of age. Of those 76 children, 29 seroconverted to T1D-related autoimmunity (cases) including 22 who later developed T1D, the remaining 47 subjects remained healthy (controls). While several significant compositional differences in low abundant species prior to seroconversion were found, one highly abundant group composed of two closely related species, Bacteroides dorei and Bacteroides vulgatus, was significantly higher in cases compared to controls prior to seroconversion. Metagenomic sequencing of samples high in the abundance of the B. dorei/vulgatus group before seroconversion, as well as longer 16S rRNA sequencing identified this group as Bacteroides dorei. The abundance of B. dorei peaked at 7.6 months in cases, over 8 months prior to the appearance of the first islet autoantibody, suggesting that early changes in the microbiome may be useful for predicting T1D autoimmunity in genetically susceptible infants. The cause of increased B. dorei abundance in cases is not known but its timing appears to coincide with the introduction of solid food.
Collapse
Affiliation(s)
- Austin G Davis-Richardson
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Alexandria N Ardissone
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Raquel Dias
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Ville Simell
- Department of Pediatrics, Turku University Hospital and University of Turku Turku, Finland
| | - Michael T Leonard
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Kaisa M Kemppainen
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Jennifer C Drew
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Desmond Schatz
- Department of Pediatrics, University of Florida Gainesville, FL, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Gainesville, FL, USA
| | - Bryan Kolaczkowski
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Jorma Ilonen
- Department of Clinical Microbiology, University of Eastern Finland Kuopio, Finland ; Immunogenetics Laboratory, University of Turku Turku, Finland
| | - Mikael Knip
- Department of Pediatrics, Children's Hospital, University of Helsinki and Helsinki University Central Hospital Helsinki, Finland ; Diabetes and Obesity Research Program, University of Helsinki Helsinki, Finland ; Department of Pediatrics, Tampere University Hospital Tampere, Finland
| | - Jorma Toppari
- Department of Pediatrics, Turku University Hospital and University of Turku Turku, Finland
| | - Noora Nurminen
- School of Medicine, University of Tampere Tampere, Finland
| | - Heikki Hyöty
- School of Medicine, University of Tampere Tampere, Finland
| | - Riitta Veijola
- Department of Pediatrics, University of Oulu, and Oulu University Hospital Oulu, Finland
| | - Tuula Simell
- Department of Pediatrics, Turku University Hospital and University of Turku Turku, Finland
| | - Juha Mykkänen
- Department of Pediatrics, Turku University Hospital and University of Turku Turku, Finland
| | - Olli Simell
- Department of Pediatrics, Turku University Hospital and University of Turku Turku, Finland
| | - Eric W Triplett
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| |
Collapse
|
1598
|
Affiliation(s)
| | - C Bradford
- Centre of Diabetes, Blizard Institute, University of London, London, U.K
| |
Collapse
|
1599
|
Drescher KM, von Herrath M, Tracy S. Enteroviruses, hygiene and type 1 diabetes: toward a preventive vaccine. Rev Med Virol 2014; 25:19-32. [PMID: 25430610 DOI: 10.1002/rmv.1815] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/26/2014] [Accepted: 10/02/2014] [Indexed: 12/24/2022]
Abstract
Enteroviruses and humans have long co-existed. Although recognized in ancient times, poliomyelitis and type 1 diabetes (T1D) were exceptionally rare and not epidemic, due in large part to poor sanitation and personal hygiene which resulted in repeated exposure to fecal-oral transmitted viruses and other infectious agents and viruses and the generation of a broad protective immunity. As a function of a growing acceptance of the benefits of hygienic practices and microbiologically clean(er) water supplies, the likelihood of exposure to diverse infectious agents and viruses declined. The effort to vaccinate against poliomyelitis demonstrated that enteroviral diseases are preventable by vaccination and led to understanding how to successfully attenuate enteroviruses. Type 1 diabetes onset has been convincingly linked to infection by numerous enteroviruses including the group B coxsackieviruses (CVB), while studies of CVB infections in NOD mice have demonstrated not only a clear link between disease onset but an ability to reduce the incidence of T1D as well: CVB infections can suppress naturally occurring autoimmune T1D. We propose here that if we can harness and develop the capacity to use attenuated enteroviral strains to induce regulatory T cell populations in the host through vaccination, then a vaccine could be considered that should function to protect against both autoimmune as well as virus-triggered T1D. Such a vaccine would not only specifically protect from certain enterovirus types but more importantly, also reset the organism's regulatory rheostat making the further development of pathogenic autoimmunity less likely.
Collapse
Affiliation(s)
- Kristen M Drescher
- Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, NE, USA
| | | | | |
Collapse
|
1600
|
Abstract
Two groups recently reported the in vitro differentiation of human embryonic stem cells into insulin-secreting cells, achieving an elusive goal for regenerative medicine. Herein we provide a perspective regarding these developments, compare phenotypes of the insulin-containing cells to human β cells, and discuss implications for type 1 diabetes research and clinical care.
Collapse
|