101
|
Burg AR, Das S, Padgett LE, Koenig ZE, Tse HM. Superoxide Production by NADPH Oxidase Intensifies Macrophage Antiviral Responses during Diabetogenic Coxsackievirus Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:61-70. [PMID: 29158420 PMCID: PMC5736405 DOI: 10.4049/jimmunol.1700478] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 10/17/2017] [Indexed: 02/07/2023]
Abstract
Coxsackievirus B infections are suspected environmental triggers of type 1 diabetes (T1D) and macrophage antiviral responses may provide a link to virus-induced T1D. We previously demonstrated an important role for NADPH oxidase (NOX)-derived superoxide production during T1D pathogenesis, as NOX-deficient NOD mice (NOD.Ncf1m1J ) were protected against T1D due, in part, to impaired proinflammatory TLR signaling in NOD.Ncf1m1J macrophages. Therefore, we hypothesized that loss of NOX-derived superoxide would dampen diabetogenic antiviral macrophage responses and protect from virus-induced diabetes. Upon infection with a suspected diabetogenic virus, Coxsackievirus B3 (CB3), NOD.Ncf1m1J mice remained resistant to virus-induced autoimmune diabetes. A concomitant decrease in circulating inflammatory chemokines, blunted antiviral gene signature within the pancreas, and reduced proinflammatory M1 macrophage responses were observed. Importantly, exogenous superoxide addition to CB3-infected NOD.Ncf1m1J bone marrow-derived macrophages rescued the inflammatory antiviral M1 macrophage response, revealing reduction-oxidation-dependent mechanisms of signal transducer and activator of transcription 1 signaling and dsRNA viral sensors in macrophages. We report that superoxide production following CB3 infection may exacerbate pancreatic β cell destruction in T1D by influencing proinflammatory M1 macrophage responses, and mechanistically linking oxidative stress, inflammation, and diabetogenic virus infections.
Collapse
Affiliation(s)
- Ashley R Burg
- Comprehensive Diabetes Center, Department of Microbiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294-2182
| | - Shaonli Das
- Comprehensive Diabetes Center, Department of Microbiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294-2182
| | - Lindsey E Padgett
- Comprehensive Diabetes Center, Department of Microbiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294-2182
| | - Zachary E Koenig
- Comprehensive Diabetes Center, Department of Microbiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294-2182
| | - Hubert M Tse
- Comprehensive Diabetes Center, Department of Microbiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294-2182
| |
Collapse
|
102
|
Jones IV AR, Coleman EL, Husni NR, Deeney JT, Raval F, Steenkamp D, Dooms H, Nikolajczyk BS, Corkey BE. Type 1 diabetes alters lipid handling and metabolism in human fibroblasts and peripheral blood mononuclear cells. PLoS One 2017; 12:e0188474. [PMID: 29206239 PMCID: PMC5714353 DOI: 10.1371/journal.pone.0188474] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 11/07/2017] [Indexed: 01/05/2023] Open
Abstract
Triggers of the autoimmune response that leads to type 1 diabetes (T1D) remain poorly understood. A possibility is that parallel changes in both T cells and target cells provoke autoimmune attack. We previously documented greater Ca2+ transients in fibroblasts from T1D subjects than non-T1D after exposure to fatty acids (FA) and tumor necrosis factor α (TNFα). These data indicate that metabolic and signal transduction defects present in T1D can be elicited ex vivo in isolated cells. Changes that precede T1D, including inflammation, may activate atypical responses in people that are genetically predisposed to T1D. To identify such cellular differences in T1D, we quantified a panel of metabolic responses in fibroblasts and peripheral blood cells (PBMCs) from age-matched T1D and non-T1D subjects, as models for non-immune and immune cells, respectively. Fibroblasts from T1D subjects accumulated more lipid, had higher LC-CoA levels and converted more FA to CO2, with less mitochondrial proton leak in response to oleate alone or with TNFα, using the latter as a model of inflammation. T1D-PBMCs contained and also accumulated more lipid following FA exposure. In addition, they formed more peroxidized lipid than controls following FA exposure. We conclude that both immune and non-immune cells in T1D subjects differ from controls in terms of responses to FA and TNFα. Our results suggest a differential sensitivity to inflammatory insults and FA that may precede and contribute to T1D by priming both immune cells and their targets for autoimmune reactions.
Collapse
Affiliation(s)
- Albert R. Jones IV
- Obesity Research Center, Evans Department of Medicine, Boston University School of Medicine, Boston, MA, United States of America
| | - Emily L. Coleman
- Yale University School of Medicine, New Haven, CT, United States of America
| | - Nicholas R. Husni
- Obesity Research Center, Evans Department of Medicine, Boston University School of Medicine, Boston, MA, United States of America
| | - Jude T. Deeney
- Obesity Research Center, Evans Department of Medicine, Boston University School of Medicine, Boston, MA, United States of America
| | - Forum Raval
- Department of Microbiology, Boston University School of Medicine, Boston, MA, United States of America
| | - Devin Steenkamp
- Endocrinology Section, Evans Department of Medicine, Boston University School of Medicine, Boston, MA, United States of America
| | - Hans Dooms
- Rheumatology Section, Evans Department of Medicine, Boston University School of Medicine, Boston, MA, United States of America
| | - Barbara S. Nikolajczyk
- Department of Translational Research in Diabetes, University of Kentucky School of Medicine, Lexington, KY, United States of America
- Department of Pharmacology and Nutritional Sciences, University of Kentucky School of Medicine, Lexington, KY, United States of America
| | - Barbara E. Corkey
- Obesity Research Center, Evans Department of Medicine, Boston University School of Medicine, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
103
|
Abstract
OBJECTIVES The aims of this study were to investigate the presence of human herpesvirus 6 (HHV6) A and B in human pancreata and to search for signs of active infection in this organ of subjects with and without type 1 diabetes (T1D). METHODS Pancreata from brain-dead organ donors with and without T1D were examined for the presence of HHV6 genomic sequences by polymerase chain reaction (PCR), transcripts by reverse transcriptase-PCR, and protein by immunohistochemistry. Quantitative PCR of isolated pancreatic islets and exocrine cell clusters was used to determine the intrapancreatic location of HHV6 DNA. RESULTS Human herpesvirus 6B genomic sequences were present in 1 of 2 donors who died of acute-onset T1D, 4 of 6 donors with long-standing T1D, and 9 of 12 nondiabetic donors. Higher copy numbers of HHV6B DNA were present in isolated islets than in exocrine tissue from the same donors. No signs of active HHV6 transcription were found. Human herpesvirus 6A was not present in any tested pancreas. CONCLUSIONS The herein presented data demonstrate, for the first time, the presence of a latent HHV6B infection in the pancreas and islets of Langerhans. Whether this virus can contribute to disease in the pancreas remains to be determined.
Collapse
|
104
|
Alidjinou EK, Sane F, Lefevre C, Baras A, Moumna I, Engelmann I, Vantyghem MC, Hober D. Enteroviruses in blood of patients with type 1 diabetes detected by integrated cell culture and reverse transcription quantitative real-time PCR. Acta Diabetol 2017; 54:1025-1029. [PMID: 28861621 DOI: 10.1007/s00592-017-1041-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/16/2017] [Indexed: 10/19/2022]
Abstract
AIMS Enteroviruses (EV) have been associated with type 1 diabetes (T1D), but EV RNA detection has been reported in only a small proportion of T1D patients. We studied whether integrated cell culture and reverse transcription real-time PCR could improve EV detection in blood samples from patients with T1D. METHODS Blood was collected from 13 patients with T1D. The presence of EV RNA in blood was investigated by using real-time RT-PCR. In addition, plasma and white blood cells (WBC) were inoculated to BGM and Vero cell line cultures. Culture supernatants and cells collected on day 7 and day 14 were tested for EV RNA by real-time RT-PCR. Enterovirus identification was performed through sequencing of the VP4/VP2 region. RESULTS Enterovirus RNA was detected in blood by using real-time RT-PCR in only one out of 13 patients. The detection of EV RNA in cultures inoculated with clinical samples (plasma and/or WBC) gave positive results in five other patients. The viral loads were low, ranging from 45 to 4420 copies/ng of total RNA. One isolate was successfully identified as coxsackievirus B1. CONCLUSIONS Integrated cell culture and reverse transcription real-time PCR can improve the detection rate of EV in blood samples of patients with T1D and can be useful to investigate further the relationship between EV and the disease.
Collapse
Affiliation(s)
- Enagnon Kazali Alidjinou
- Laboratoire de Virologie EA3610, Univ Lille, Faculté de Médecine, CHU Lille, 59000, Lille, France
| | - Famara Sane
- Laboratoire de Virologie EA3610, Univ Lille, Faculté de Médecine, CHU Lille, 59000, Lille, France
| | - Christine Lefevre
- Service d'Endocrinologie pédiatrique, CHU Lille, 59000, Lille, France
| | - Agathe Baras
- Laboratoire de Virologie EA3610, Univ Lille, Faculté de Médecine, CHU Lille, 59000, Lille, France
| | - Ilham Moumna
- Laboratoire de Virologie EA3610, Univ Lille, Faculté de Médecine, CHU Lille, 59000, Lille, France
| | - Ilka Engelmann
- Laboratoire de Virologie EA3610, Univ Lille, Faculté de Médecine, CHU Lille, 59000, Lille, France
| | | | - Didier Hober
- Laboratoire de Virologie EA3610, Univ Lille, Faculté de Médecine, CHU Lille, 59000, Lille, France.
| |
Collapse
|
105
|
Badia-Boungou F, Sane F, Alidjinou EK, Ternois M, Opoko PA, Haddad J, Stukens C, Lefevre C, Gueorguieva I, Hamze M, Ismail M, Weill J, Monabéka HG, Bouenizabila E, Moukassa D, Abena AA, Hober D. Marker of coxsackievirus-B4 infection in saliva of patients with type 1 diabetes. Diabetes Metab Res Rev 2017; 33. [PMID: 28719027 DOI: 10.1002/dmrr.2916] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 06/20/2017] [Accepted: 07/03/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Coxsackieviruses B (CV-B) are enteroviruses that have been reported to play a role in the pathogenesis of type 1 diabetes. Enteroviral RNA was detected in the gut mucosa of patients. The mucosal immunity is an interconnected network; therefore, the response to enteroviruses possibly present in the gastrointestinal mucosa can be reflected by specific antibodies in the saliva. In the present study, the anti-CV-B neutralizing activity of saliva samples from patients with type 1 diabetes was investigated. METHODS Saliva samples were collected from patients and controls of 3 countries, and plasma was obtained from some of them. The anti-CV-B activity of clinical samples was determined by neutralization of the cytopathic effect induced by challenging viruses in vitro and expressed as titre value. RESULTS Overall prevalence and levels of anti-CV-B4 activity of saliva were higher in patients (n = 181) than in controls (n = 135; P = .0002; titre values ≥ 16: odds ratio = 4.22 95% CI: 1.90-9.38 P = .0002). It has been shown that IgA1 played a role in this activity. There was no correlation between the saliva and the plasma anti-CV-B4 neutralizing activity. The neutralizing activity of saliva against CV-B1, CV-B2, CV-B3, and CV-B5 existed rarely, if at all. Increased levels of anti-CV-B4 activity were observed all along a 4 year follow-up period in patients but not in matched controls (P = .01). CONCLUSION There is an anti-CV-B4 activity in saliva of patients with type 1 diabetes that may be a useful marker to study the role of CV-B in the pathogenesis of the disease.
Collapse
Affiliation(s)
- F Badia-Boungou
- Univ. Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, France
- Hôpital Général de Loandjili, Pointe Noire, Republic of Congo
| | - F Sane
- Univ. Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, France
| | - E K Alidjinou
- Univ. Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, France
| | - M Ternois
- Univ. Lille, Faculté d'odontologie et de chirurgie dentaire, Lille, France
| | - P A Opoko
- Hôpital Général de Loandjili, Pointe Noire, Republic of Congo
| | - J Haddad
- Lebanese University Faculty of Public Health, Health and Environment Microbiology Laboratory, Tripoli, Lebanon
| | - C Stukens
- CHU Hôpital Jeanne de Flandres, Lille, France
| | - C Lefevre
- CHU Hôpital Jeanne de Flandres, Lille, France
| | | | - M Hamze
- Lebanese University Faculty of Public Health, Health and Environment Microbiology Laboratory, Tripoli, Lebanon
| | - M Ismail
- Lebanese University Faculty of Public Health, Health and Environment Microbiology Laboratory, Tripoli, Lebanon
| | - J Weill
- CHU Hôpital Jeanne de Flandres, Lille, France
| | - H G Monabéka
- Université Marien Ngouabi, Faculté des sciences et de la santé, Brazzaville, République du Congo
- CHU de Brazzaville, Brazzaville, Republic of Congo
| | | | - D Moukassa
- Hôpital Général de Loandjili, Pointe Noire, Republic of Congo
- Université Marien Ngouabi, Faculté des sciences et de la santé, Brazzaville, République du Congo
| | - A A Abena
- Université Marien Ngouabi, Faculté des sciences et de la santé, Brazzaville, République du Congo
| | - D Hober
- Univ. Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, France
| |
Collapse
|
106
|
Lönnrot M, Lynch KF, Elding Larsson H, Lernmark Å, Rewers MJ, Törn C, Burkhardt BR, Briese T, Hagopian WA, She JX, Simell OG, Toppari J, Ziegler AG, Akolkar B, Krischer JP, Hyöty H. Respiratory infections are temporally associated with initiation of type 1 diabetes autoimmunity: the TEDDY study. Diabetologia 2017; 60:1931-1940. [PMID: 28770319 PMCID: PMC5697762 DOI: 10.1007/s00125-017-4365-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 06/05/2017] [Indexed: 12/15/2022]
Abstract
AIMS/HYPOTHESIS Respiratory infections and onset of islet autoimmunity are reported to correlate positively in two small prospective studies. The Environmental Determinants of Diabetes in the Young (TEDDY) study is the largest prospective international cohort study on the environmental determinants of type 1 diabetes that regularly monitors both clinical infections and islet autoantibodies. The aim was to confirm the influence of reported respiratory infections and to further characterise the temporal relationship with autoantibody seroconversion. METHODS During the years 2004-2009, 8676 newborn babies with HLA genotypes conferring an increased risk of type 1 diabetes were enrolled at 3 months of age to participate in a 15 year follow-up. In the present study, the association between parent-reported respiratory infections and islet autoantibodies at 3 month intervals up to 4 years of age was evaluated in 7869 children. Time-dependent proportional hazard models were used to assess how the timing of respiratory infections related to persistent confirmed islet autoimmunity, defined as autoantibody positivity against insulin, GAD and/or insulinoma antigen-2, concordant at two reference laboratories on two or more consecutive visits. RESULTS In total, 87,327 parent-reported respiratory infectious episodes were recorded while the children were under study surveillance for islet autoimmunity, and 454 children seroconverted. The number of respiratory infections occurring in a 9 month period was associated with the subsequent risk of autoimmunity (p < 0.001). For each 1/year rate increase in infections, the hazard of islet autoimmunity increased by 5.6% (95% CI 2.5%, 8.8%). The risk association was linked primarily to infections occurring in the winter (HR 1.42 [95% CI 1.16, 1.74]; p < 0.001). The types of respiratory infection independently associated with autoimmunity were common cold, influenza-like illness, sinusitis, and laryngitis/tracheitis, with HRs (95% CI) of 1.38 (1.11, 1.71), 2.37 (1.35, 4.15), 2.63 (1.22, 5.67) and 1.76 (1.04, 2.98), respectively. CONCLUSIONS/INTERPRETATION Recent respiratory infections in young children correlate with an increased risk of islet autoimmunity in the TEDDY study. Further studies to identify the potential causative viruses with pathogen-specific assays should focus especially on the 9 month time window leading to autoantibody seroconversion.
Collapse
Affiliation(s)
- Maria Lönnrot
- Department of Dermatology, Tampere University Hospital, Teiskontie 35, 33521, Tampere, Finland.
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.
| | - Kristian F Lynch
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Helena Elding Larsson
- Department of Clinical Sciences Malmö, Clinical Research Centre (CRC), Lund University, Malmö, Sweden
- Skåne University Hospital (SUS), Malmo, Sweden
| | - Åke Lernmark
- Skåne University Hospital (SUS), Malmo, Sweden
- Department of Clinical Sciences Malmö, Lund University Clinical Research Centre (CRC), Malmö, Sweden
| | - Marian J Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Carina Törn
- Department of Clinical Sciences Malmö, Clinical Research Centre (CRC), Lund University, Malmö, Sweden
- Skåne University Hospital (SUS), Malmo, Sweden
| | - Brant R Burkhardt
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Thomas Briese
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | | | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Olli G Simell
- Research Centre of Applied and Preventive Cardiovascular Medicine, Faculty of Medicine, University of Turku, Turku, Finland
- Department of Paediatrics and Adolescent Medicine, Faculty of Medicine, University of Turku, Turku, Finland
| | - Jorma Toppari
- Department of Physiology, Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Paediatrics, Turku University Hospital, Turku, Finland
| | - Anette-G Ziegler
- Forschergruppe Diabetes e.V, Neuherberg, Germany
- Institute of Diabetes Research, Helmholtz Zentrum München, Munich, Germany
| | - Beena Akolkar
- National Institute of Diabetes & Digestive & Kidney Diseases, Bethesda, MD, USA
| | - Jeffrey P Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Heikki Hyöty
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| |
Collapse
|
107
|
Abstract
PURPOSE OF REVIEW Despite immense research efforts, type 1 diabetes (T1D) remains an autoimmune disease without a known trigger or approved intervention. Over the last three decades, studies have primarily focused on delineating the role of the adaptive immune system in the mechanism of T1D. The discovery of Toll-like receptors in the 1990s has advanced the knowledge on the role of the innate immune system in host defense as well as mechanisms that regulate adaptive immunity including the function of autoreactive T cells. RECENT FINDINGS Recent investigations suggest that inflammation plays a key role in promoting a large number of autoimmune disorders including T1D. Data from the LEW1.WR1 rat model of virus-induced disease and the RIP-B7.1 mouse model of diabetes suggest that innate immune signaling plays a key role in triggering disease progression. There is also evidence that innate immunity may be involved in the course of T1D in humans; however, a small number of clinical trials have shown that interfering with the function of the innate immune system following disease onset exerts only a modest effect on β-cell function. The data implying that innate immune pathways are linked with mechanisms of islet autoimmunity hold great promise for the identification of novel disease pathways that may be harnessed for clinical intervention. Nevertheless, more work needs to be done to better understand mechanisms by which innate immunity triggers β-cell destruction and assess the therapeutic value in blocking innate immunity for diabetes prevention.
Collapse
Affiliation(s)
- James C Needell
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Danny Zipris
- Innate Biotechnologies LLC, Denver, CO, 80231, USA.
| |
Collapse
|
108
|
Semeraro ML, Glenn LM, Morris MA. The Four-Way Stop Sign: Viruses, 12-Lipoxygenase, Islets, and Natural Killer Cells in Type 1 Diabetes Progression. Front Endocrinol (Lausanne) 2017; 8:246. [PMID: 28993759 PMCID: PMC5622285 DOI: 10.3389/fendo.2017.00246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/08/2017] [Indexed: 12/29/2022] Open
Abstract
Natural killer (NK) cells represent an important effector arm against viral infection, and mounting evidence suggests that viral infection plays a role in the development of type 1 diabetes (T1D) in at least a portion of patients. NK cells recognize their target cells through a delicate balance of inhibitory and stimulatory receptors on their surface. If unbalanced, NK cells have great potential to wreak havoc in the pancreas due to the beta cell expression of the as-yet-defined NKp46 ligand through interactions with the activating NKp46 receptor found on the surface of most NK cells. Blocking interactions between NKp46 and its ligand protects mice from STZ-induced diabetes, but differential expression non-diabetic and diabetic donor samples have not been tested. Additional studies have shown that peripheral blood NK cells from human T1D patients have altered phenotypes that reduce the lytic and functional ability of the NK cells. Investigations of humanT1D pancreas tissues have indicated that the presence of NK cells may be beneficial despite their infrequent detection. In non-obese diabetic (NOD) mice, we have noted that NK cells express high levels of the proinflammatory mediator 12/15-lipoxygenase (12/15-LO), and decreased levels of stimulatory receptors. Conversely, NK cells of 12/15-LO deficient NOD mice, which are protected from diabetes development, express significantly higher levels of stimulatory receptors. Furthermore, the human NK92 cell line expresses the ALOX12 protein [human 12-lipoxygenase (12-LO), related to mouse 12/15-LO] via Western blotting. Human 12-LO is upregulated in the pancreas of both T1D and T2D human donors with insulin-containing islets, showing a link between 12-LO expression and diabetes progression. Therefore, our hypothesis is that NK cells in those susceptible to developing T1D are unable to function properly during viral infections of pancreatic beta cells due to increased 12-LO expression and activation, which contributes to increased interferon-gamma production and an imbalance in activating and inhibitory NK cell receptors, and may contribute to downstream autoimmune T cell responses. The work presented here outlines evidence from our lab, as well as published literature, supporting our hypothesis, including novel data.
Collapse
Affiliation(s)
- Michele L. Semeraro
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Lindsey M. Glenn
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Margaret A. Morris
- Department of Internal Medicine, Strelitz Diabetes Center, Eastern Virginia Medical School, Norfolk, VA, United States
| |
Collapse
|
109
|
Busse N, Paroni F, Richardson SJ, Laiho JE, Oikarinen M, Frisk G, Hyöty H, de Koning E, Morgan NG, Maedler K. Detection and localization of viral infection in the pancreas of patients with type 1 diabetes using short fluorescently-labelled oligonucleotide probes. Oncotarget 2017; 8:12620-12636. [PMID: 28147344 PMCID: PMC5355040 DOI: 10.18632/oncotarget.14896] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/19/2017] [Indexed: 01/08/2023] Open
Abstract
Enteroviruses, specifically of the Coxsackie B virus family, have been implicated in triggering islet autoimmunity and type 1 diabetes, but their presence in pancreata of patients with diabetes has not been fully confirmed. To detect the presence of very low copies of the virus genome in tissue samples from T1D patients, we designed a panel of fluorescently labeled oligonucleotide probes, each of 17-22 nucleotides in length with a unique sequence to specifically bind to the enteroviral genome of the picornaviridae family. With these probes enteroviral RNA was detected with high sensitivity and specificity in infected cells and tissues, including in FFPE pancreas sections from patients with T1D. Detection was not impeded by variations in sample processing and storage thereby overcoming the potential limitations of fragmented RNA. Co-staining of small RNA probes in parallel with classical immunstaining enabled virus detection in a cell-specific manner and more sensitively than by viral protein.
Collapse
Affiliation(s)
- Niels Busse
- Islet Biology Laboratory, University of Bremen, Germany
| | | | | | - Jutta E Laiho
- Department of Virology, School of Medicine, University of Tampere, Tampere, Finland
| | - Maarit Oikarinen
- Department of Virology, School of Medicine, University of Tampere, Tampere, Finland
| | - Gun Frisk
- Department of Immunology, Genetics and Pathology, Uppsala University, Sweden
| | - Heikki Hyöty
- Department of Virology, School of Medicine, University of Tampere, Tampere, Finland.,Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| | - Eelco de Koning
- Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Hubrecht Institute/University Medical Center Utrecht, Utrecht, The Netherlands
| | - Noel G Morgan
- Islet Biology Exeter, University of Exeter Medical School, UK
| | | |
Collapse
|
110
|
Dou Y, Yim HC, Kirkwood CD, Williams BR, Sadler AJ. The innate immune receptor MDA5 limits rotavirus infection but promotes cell death and pancreatic inflammation. EMBO J 2017; 36:2742-2757. [PMID: 28851763 PMCID: PMC5599799 DOI: 10.15252/embj.201696273] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 07/25/2017] [Accepted: 07/28/2017] [Indexed: 12/31/2022] Open
Abstract
Melanoma differentiation-associated protein 5 (MDA5) mediates the innate immune response to viral infection. Polymorphisms in IFIH1, the gene coding for MDA5, correlate with the risk of developing type 1 diabetes (T1D). Here, we demonstrate that MDA5 is crucial for the immune response to enteric rotavirus infection, a proposed etiological agent for T1D. MDA5 variants encoded by minor IFIH1 alleles associated with lower T1D risk exhibit reduced activity against rotavirus infection. We find that MDA5 activity limits rotavirus infection not only through the induction of antiviral interferons and pro-inflammatory cytokines, but also by promoting cell death. Importantly, this MDA5-dependent antiviral response is specific to the pancreas of rotavirus-infected mice, similar to the autoimmunity associated with T1D. These findings imply that MDA5-induced cell death and inflammation in the pancreas facilitate progression to autoimmune destruction of pancreatic β-cells.
Collapse
Affiliation(s)
- Yu Dou
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Vic., Australia
- Department of Oral and Maxillofacial Surgery, Institute of Dental Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Howard Ch Yim
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Vic., Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Vic., Australia
| | - Carl D Kirkwood
- Enteric and Diarrheal Disease, Global Health, Bill and Melinda Gates Foundation, Seattle, WA, USA
- Murdoch Childrens Research Institute, The Royal Children's Hospital, Parkville, Vic., Australia
| | - Bryan Rg Williams
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Vic., Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Vic., Australia
| | - Anthony J Sadler
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Vic., Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Vic., Australia
| |
Collapse
|
111
|
Principi N, Berioli MG, Bianchini S, Esposito S. Type 1 diabetes and viral infections: What is the relationship? J Clin Virol 2017; 96:26-31. [PMID: 28934695 DOI: 10.1016/j.jcv.2017.09.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 08/09/2017] [Accepted: 09/06/2017] [Indexed: 12/16/2022]
Abstract
Type 1 diabetes (T1D) is the most common chronic metabolic disorder in children. Epigenetic and environmental factors capable of altering the penetrance of major susceptibility genes or capable of increasing the penetrance of low-risk genes are currently thought to play a role in triggering autoimmunity and T1D development. This paper discusses the current knowledge of the role of viruses in T1D. Most studies that have evaluated the potential association between viral infections and T1D have indicated that it is highly likely that some of these infectious agents play a role in T1D development. However, most T1D cases are immune-mediated, and it is supposed that the initial viral infection is capable of creating, in genetically predisposed subjects, a particular condition in which chronic local inflammation occurs through the persistence of the infecting virus in pancreatic tissue and the activation of autoimmunity by means of molecular mimicry, bystander activation, or both. Theoretically, this knowledge could lead to possible prophylaxis and therapy for T1D. Further studies devoted to evaluating which infectious agents are linked to T1D and which immune mechanisms induce or protect against the disease are needed before adequate prophylactic and therapeutic measures can be developed.
Collapse
Affiliation(s)
- Nicola Principi
- Professor Emeritus, Università degli Studi di Milano, Milan, Italy
| | | | - Sonia Bianchini
- Pediatric Clinic, Università degli Studi di Perugia, Perugia, Italy
| | - Susanna Esposito
- Pediatric Clinic, Università degli Studi di Perugia, Perugia, Italy.
| |
Collapse
|
112
|
Nasir IA, Emeribe AU, Shuwa HA, Zakari MM, Peters NO. Type 1 diabetes mellitus and enterovirus linkage: search for associated etiopathology. THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2017. [DOI: 10.4103/ejim.ejim_25_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
113
|
Elmastour F, Jaïdane H, Benkahla M, Aguech-Oueslati L, Sane F, Halouani A, Engelmann I, Bertin A, Mokni M, Gharbi J, Aouni M, Alidjinou EK, Hober D. Anti-coxsackievirus B4 (CV-B4) enhancing activity of serum associated with increased viral load and pathology in mice reinfected with CV-B4. Virulence 2017; 8:908-923. [PMID: 27792461 PMCID: PMC5626334 DOI: 10.1080/21505594.2016.1252018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 09/29/2016] [Accepted: 10/18/2016] [Indexed: 12/15/2022] Open
Abstract
In previous studies it was shown that inoculation of Swiss albino mice with CV-B4 E2 resulted in the production of serum IgG capable of enhancing the CV-B4 E2 infection of murine spleen cells cultures. To investigate whether such an enhancing activity of serum can play a role in vivo, we decided to study the CV-B4 E2 infection in mice exposed to successive inoculations of virus. In Swiss albino mice infected with CV-B4 E2 at the age of 21 days, anti-CV-B4 E2 neutralizing and enhancing activities of their serum peaked after 55 d. In contrast, mice inoculated at the age of 55 d expressed much lower activities. Despite the neutralizing activity of serum, CV-B4 E2 inoculated a second time to 55 day-old animals spread into the host. At the age of 72 and 89 d the levels of viral RNA and infectious particles were higher in organs of animals exposed to 2 successive infections compared with animals infected once at the age of 21 d or 55 d. In animals with 2 successive inoculations of CV-B4 E2 there was a relationship between the anti-CV-B4 E2 enhancing activity of serum and the level of viral RNA in organs and an enhancement of pathology was observed as displayed by histological analysis of pancreas and hyperglycaemia. Altogether our data strongly suggest that an anti-CV-B4 E2 enhancing activity in the host can play a role in the outcome of a secondary infection with this virus.
Collapse
Affiliation(s)
- Firas Elmastour
- Université de Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie/EA3610, Lille, France
- Université de Monastir, Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir, Monastir, Tunisia
- Université de Tunis El Manar, Faculté des Sciences de Tunis, Tunis, Tunisia
| | - Hela Jaïdane
- Université de Monastir, Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir, Monastir, Tunisia
- Université de Tunis El Manar, Faculté des Sciences de Tunis, Tunis, Tunisia
| | - Mehdi Benkahla
- Université de Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie/EA3610, Lille, France
| | - Leila Aguech-Oueslati
- Université de Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie/EA3610, Lille, France
- Université de Monastir, Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir, Monastir, Tunisia
| | - Famara Sane
- Université de Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie/EA3610, Lille, France
| | - Aymen Halouani
- Université de Monastir, Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir, Monastir, Tunisia
- Université de Tunis El Manar, Faculté des Sciences de Tunis, Tunis, Tunisia
| | - Ilka Engelmann
- Université de Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie/EA3610, Lille, France
| | - Antoine Bertin
- Université de Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie/EA3610, Lille, France
| | - Moncef Mokni
- Université de Sousse, CHU Farhat Hached, Service d'Anatomopathologie, Sousse, Tunisia
| | - Jawhar Gharbi
- Université de Monastir, Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir, Monastir, Tunisia
| | - Mahjoub Aouni
- Université de Monastir, Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir, Monastir, Tunisia
| | - Enagnon K. Alidjinou
- Université de Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie/EA3610, Lille, France
| | - Didier Hober
- Université de Lille, Faculté de Médecine, CHU Lille, Laboratoire de Virologie/EA3610, Lille, France
| |
Collapse
|
114
|
Ma WT, Chang C, Gershwin ME, Lian ZX. Development of autoantibodies precedes clinical manifestations of autoimmune diseases: A comprehensive review. J Autoimmun 2017; 83:95-112. [PMID: 28739356 DOI: 10.1016/j.jaut.2017.07.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/30/2017] [Accepted: 07/01/2017] [Indexed: 12/21/2022]
Abstract
The etiology of autoimmune diseases is due to a combination of genetic predisposition and environmental factors that alter the expression of immune regulatory genes through various mechanisms including epigenetics. Both humoral and cellular elements of the adaptive immune system play a role in the pathogenesis of autoimmune diseases and the presence of autoantibodies have been detected in most but not all autoimmune diseases before the appearance of clinical symptoms. In some cases, the presence or levels of these autoantibodies portends not only the risk of developing a corresponding autoimmune disease, but occasionally the severity as well. This observation is intriguing because it suggests that we can, to some degree, predict who may or may not develop autoimmune diseases. However, the role of autoantibodies in the pathogenesis of autoimmune diseases, whether they actually affect disease progression or are merely an epiphenomenon is still not completely clear in many autoimmune diseases. Because of these gaps in our knowledge, the ability to accurately predict a future autoimmune disease can only be considered a relative risk factor. Importantly, it raises the critical question of defining other events that may drive a patient from a preclinical to a clinical phase of disease.
Collapse
Affiliation(s)
- Wen-Tao Ma
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou 510006, China; Liver Immunology Laboratory, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China; College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling 712100, China
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA, USA
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA, USA.
| | - Zhe-Xiong Lian
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou 510006, China; Liver Immunology Laboratory, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China; Innovation Center for Cell Signaling Network, Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui, China.
| |
Collapse
|
115
|
Willcox A, Richardson SJ, Walker LSK, Kent SC, Morgan NG, Gillespie KM. Germinal centre frequency is decreased in pancreatic lymph nodes from individuals with recent-onset type 1 diabetes. Diabetologia 2017; 60:1294-1303. [PMID: 28213757 PMCID: PMC5487689 DOI: 10.1007/s00125-017-4221-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 12/13/2016] [Indexed: 01/11/2023]
Abstract
AIMS/HYPOTHESIS Pancreatic lymph nodes (PLNs) are critical sites for the initial interaction between islet autoantigens and autoreactive lymphocytes, but the histology of PLNs in tissue from individuals with type 1 diabetes has not been analysed in detail. The aim of this study was to examine PLN tissue sections from healthy donors compared with those at risk of, or with recent-onset and longer-duration type 1 diabetes. METHODS Immunofluorescence staining was used to examine PLN sections from the following donor groups: non-diabetic (n=15), non-diabetic islet autoantibody-positive (n=5), recent-onset (≤1.5 years duration) type 1 diabetes (n=13), and longer-duration type 1 diabetes (n=15). Staining for CD3, CD20 and Ki67 was used to detect primary and secondary (germinal centre-containing) follicles and CD21 and CD35 to detect follicular dendritic cell networks. RESULTS The frequency of secondary follicles was lower in the recent-onset type 1 diabetes group compared with the non-diabetic control group. The presence of insulitis (as evidence of ongoing beta cell destruction) and diagnosis of type 1 diabetes at a younger age, however, did not appear to be associated with a lower frequency of secondary follicles. A higher proportion of primary B cell follicles were observed to lack follicular dendritic cell networks in the recent-onset type 1 diabetes group. CONCLUSIONS/INTERPRETATION Histological analysis of rare PLNs from individuals with type 1 diabetes suggests a previously unrecognised phenotype comprising decreased primary B cell follicle frequency and fewer follicular dendritic cell networks in recent-onset type 1 diabetes.
Collapse
Affiliation(s)
- Abby Willcox
- Diabetes and Metabolism, Level 2 Learning and Research, University of Bristol, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Sarah J Richardson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Lucy S K Walker
- Institute of Immunity and Transplantation, UCL Division of Infection and Immunity, Royal Free Campus, London, UK
| | - Sally C Kent
- Division of Diabetes, Department of Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA, USA
| | - Noel G Morgan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Kathleen M Gillespie
- Diabetes and Metabolism, Level 2 Learning and Research, University of Bristol, Southmead Hospital, Bristol, BS10 5NB, UK.
| |
Collapse
|
116
|
El Kfoury KA, Romond MB, Scuotto A, Alidjinou EK, Dabboussi F, Hamze M, Engelmann I, Sane F, Hober D. Bifidobacteria-derived lipoproteins inhibit infection with coxsackievirus B4 in vitro. Int J Antimicrob Agents 2017; 50:177-185. [PMID: 28595938 DOI: 10.1016/j.ijantimicag.2017.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 02/07/2017] [Accepted: 03/11/2017] [Indexed: 10/19/2022]
Abstract
The aim of the present study was to investigate the potential of bifidobacteria in protecting cells from coxsackievirus B4 (CV-B4) infection. Bifidobacterial screening identified two of five strains that protected human epithelial type 2 (HEp-2) cell viability when bifidobacteria were incubated with viral particles prior to inoculation. In contrast, no effect was shown by incubating HEp-2 cells with bifidobacteria prior to CV-B4 inoculation. Cell wall lipoprotein aggregates (LpAs) secreted by the selected strains were assayed for their antiviral activity. The two LpAs exhibited antiviral activity when they were incubated with viral particles prior to inoculation of HEp-2 cells. Recombinant LpA-derived protein exhibited identical antiviral activity. To identify the peptide sequences interacting with the virus particles, LpA proteins were aligned with the peptide sequences of the north canyon rim and puff footprint onto coxsackievirus and adenovirus receptor (CAR). The in silico molecular docking study using CV-B3 as template showed low-energy binding, indicating a stable system for the selected peptides and consequently a likely binding interaction with CV-B. Bifidobacterium longum and Bifidobacterium breve peptides homologous to the viral north rim footprint onto CAR sequence formed hydrogen bonds with several viral residues in the north rim of the canyon, which were already predicted as interacting with CAR. In conclusion, proteins from bifidobacterial LpAs can inhibit infection with CV-B4, likely through binding to the capsid amino acids that interact with CAR.
Collapse
Affiliation(s)
- Khalil Antoine El Kfoury
- Université de Lille, CHU Lille, Laboratoire de Virologie EA3610, Lille F-59000, France; Université Libanaise, Laboratoire de Microbiologie Santé et Environnement, Ecole Doctorale des Sciences et Technologie, Faculté de Santé Publique, Tripoli, Lebanon
| | | | - Angelo Scuotto
- Bifinove, 99 rue Jardin des Plantes, Lille 59000, France
| | | | - Fouad Dabboussi
- Université Libanaise, Laboratoire de Microbiologie Santé et Environnement, Ecole Doctorale des Sciences et Technologie, Faculté de Santé Publique, Tripoli, Lebanon
| | - Monzer Hamze
- Université Libanaise, Laboratoire de Microbiologie Santé et Environnement, Ecole Doctorale des Sciences et Technologie, Faculté de Santé Publique, Tripoli, Lebanon
| | - Ilka Engelmann
- Université de Lille, CHU Lille, Laboratoire de Virologie EA3610, Lille F-59000, France
| | - Famara Sane
- Université de Lille, CHU Lille, Laboratoire de Virologie EA3610, Lille F-59000, France
| | - Didier Hober
- Université de Lille, CHU Lille, Laboratoire de Virologie EA3610, Lille F-59000, France.
| |
Collapse
|
117
|
Siglec-7 restores β-cell function and survival and reduces inflammation in pancreatic islets from patients with diabetes. Sci Rep 2017; 7:45319. [PMID: 28378743 PMCID: PMC5381285 DOI: 10.1038/srep45319] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/22/2017] [Indexed: 01/16/2023] Open
Abstract
Chronic inflammation plays a key role in both type 1 and type 2 diabetes. Cytokine and chemokine production within the islets in a diabetic milieu results in β-cell failure and diabetes progression. Identification of targets, which both prevent macrophage activation and infiltration into islets and restore β-cell functionality is essential for effective diabetes therapy. We report that certain Sialic-acid-binding immunoglobulin-like-lectins (siglecs) are expressed in human pancreatic islets in a cell-type specific manner. Siglec-7 was expressed on β-cells and down-regulated in type 1 and type 2 diabetes and in infiltrating activated immune cells. Over-expression of Siglec-7 in diabetic islets reduced cytokines, prevented β-cell dysfunction and apoptosis and reduced recruiting of migrating monocytes. Our data suggest that restoration of human Siglec-7 expression may be a novel therapeutic strategy targeted to both inhibition of immune activation and preservation of β-cell function and survival.
Collapse
|
118
|
Honkanen H, Oikarinen S, Nurminen N, Laitinen OH, Huhtala H, Lehtonen J, Ruokoranta T, Hankaniemi MM, Lecouturier V, Almond JW, Tauriainen S, Simell O, Ilonen J, Veijola R, Viskari H, Knip M, Hyöty H. Detection of enteroviruses in stools precedes islet autoimmunity by several months: possible evidence for slowly operating mechanisms in virus-induced autoimmunity. Diabetologia 2017; 60:424-431. [PMID: 28070615 DOI: 10.1007/s00125-016-4177-z] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/14/2016] [Indexed: 12/15/2022]
Abstract
AIMS/HYPOTHESIS This case-control study was nested in a prospective birth cohort to evaluate whether the presence of enteroviruses in stools was associated with the appearance of islet autoimmunity in the Type 1 Diabetes Prediction and Prevention study in Finland. METHODS Altogether, 1673 longitudinal stool samples from 129 case children who turned positive for multiple islet autoantibodies and 3108 stool samples from 282 matched control children were screened for the presence of enterovirus RNA using RT-PCR. Viral genotype was detected by sequencing. RESULTS Case children had more enterovirus infections than control children (0.8 vs 0.6 infections per child). Time-dependent analysis indicated that this excess of infections occurred more than 1 year before the first detection of islet autoantibodies (6.3 vs 2.1 infections per 10 follow-up years). No such difference was seen in infections occurring less than 1 year before islet autoantibody seroconversion or after seroconversion. The most frequent enterovirus types included coxsackievirus A4 (28% of genotyped viruses), coxsackievirus A2 (14%) and coxsackievirus A16 (11%). CONCLUSIONS/INTERPRETATION The results suggest that enterovirus infections diagnosed by detecting viral RNA in stools are associated with the development of islet autoimmunity with a time lag of several months.
Collapse
Affiliation(s)
- Hanna Honkanen
- Department of Virology, University of Tampere, PL100, 33014, Tampereen yliopisto, Finland.
| | - Sami Oikarinen
- Department of Virology, University of Tampere, PL100, 33014, Tampereen yliopisto, Finland
- Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| | - Noora Nurminen
- Department of Virology, University of Tampere, PL100, 33014, Tampereen yliopisto, Finland
| | - Olli H Laitinen
- Vactech Ltd, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | - Heini Huhtala
- School of Health Sciences, University of Tampere, Tampere, Finland
| | - Jussi Lehtonen
- Department of Virology, University of Tampere, PL100, 33014, Tampereen yliopisto, Finland
| | | | - Minna M Hankaniemi
- Vactech Ltd, Tampere, Finland
- BioMediTech, University of Tampere, Tampere, Finland
| | | | | | | | - Olli Simell
- Department of Pediatrics and Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Riitta Veijola
- Department of Pediatrics, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Hanna Viskari
- Department of Virology, University of Tampere, PL100, 33014, Tampereen yliopisto, Finland
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Mikael Knip
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
- Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Heikki Hyöty
- Department of Virology, University of Tampere, PL100, 33014, Tampereen yliopisto, Finland
- Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| |
Collapse
|
119
|
Engelmann I, Dewilde A, Lazrek M, Batteux M, Hamissi A, Yakoub-Agha I, Hober D. In Vivo Persistence of Human Rhinoviruses in Immunosuppressed Patients. PLoS One 2017; 12:e0170774. [PMID: 28151988 PMCID: PMC5289482 DOI: 10.1371/journal.pone.0170774] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/10/2017] [Indexed: 12/17/2022] Open
Abstract
Several species of the genus Enterovirus cause persistent infections in humans. Human rhinovirus (HRV) infections are generally self-limiting but occasionally persistent infections have been described. This study aimed to identify persistent HRV infections and investigate the clinical and virologic characteristics of patients with persistent infections. From January 2012 to March 2015, 3714 respiratory specimens from 2608 patients were tested for respiratory viruses by using a multiplex reverse transcription–polymerase chain reaction. A retrospective study was performed. Patients with at least two specimens positive for HRV/enterovirus taken 45 days or longer apart were identified and the HRV/enteroviruses were typed. Patients with persistent infection were compared to patients with reinfection and patients with cleared infection. Phylogenetic analysis of the viral protein(VP)4/VP2 region was performed. 18 patients with persistent HRV/enterovirus infection were identified. Minimum median duration of persistence was 92 days (range 50–455 days). All but one patients with persistence were immunosuppressed. Immunosuppression and hematologic disorders were more frequent in patients with persistence (n = 18) than in patients with reinfection (n = 33) and with cleared infection (n = 25) (p = 0.003 and p = 0.001, respectively). In conclusion, this retrospective study identified HRV persistence in vivo which occurred mainly in immunosuppressed patients.
Collapse
Affiliation(s)
- Ilka Engelmann
- Laboratoire de Virologie EA 3610, Faculté de Médecine, Université Lille et CHU Lille, Lille, France
- * E-mail:
| | - Anny Dewilde
- Laboratoire de Virologie EA 3610, Faculté de Médecine, Université Lille et CHU Lille, Lille, France
| | - Mouna Lazrek
- Laboratoire de Virologie EA 3610, Faculté de Médecine, Université Lille et CHU Lille, Lille, France
| | - Mathilde Batteux
- Laboratoire de Virologie EA 3610, Faculté de Médecine, Université Lille et CHU Lille, Lille, France
| | - Aminati Hamissi
- Laboratoire de Virologie EA 3610, Faculté de Médecine, Université Lille et CHU Lille, Lille, France
| | - Ibrahim Yakoub-Agha
- Maladies du Sang, CHU Lille et Faculté de Médecine, Université Lille, Lille, France
- INSERM U995, LIRIC, Lille, France
| | - Didier Hober
- Laboratoire de Virologie EA 3610, Faculté de Médecine, Université Lille et CHU Lille, Lille, France
| |
Collapse
|
120
|
Vorobjova T, Raikkerus H, Kadaja L, Talja I, Uibo O, Heilman K, Uibo R. Circulating Zonulin Correlates with Density of Enteroviruses and Tolerogenic Dendritic Cells in the Small Bowel Mucosa of Celiac Disease Patients. Dig Dis Sci 2017; 62:358-371. [PMID: 27995404 DOI: 10.1007/s10620-016-4403-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/29/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Impaired intestinal integrity, including increased permeability of the small bowel mucosa, has been shown in patients with celiac disease (CD) as well as with type 1 diabetes (T1D). Zonulin (ZO, pre-haptoglobin), a tight junction regulator, plays a particular role in the regulation of intestinal barrier function and in the pathogenesis of the above-mentioned diseases. AIM To investigate whether enteroviruses (EVs) and immunoregulatory cells are associated with intestinal permeability in patients with CD alone and with coexistent T1D. MATERIALS AND METHODS Altogether 80 patients (mean age 10.68 ± 6.69 years) who had undergone small bowel biopsy were studied. Forty patients with functional dyspepsia and normal small bowel mucosa formed the control group. The circulating ZO level in sera was evaluated using ELISA. The densities of EV, FOXP3+ regulatory T cells (Tregs), indoleamine 2,3-dioxygenase (IDO+) dendritic cells (DCs) and glutamic acid dexarboxylase (GAD)65+ cells in small bowel mucosa were investigated by immunohistochemistry. The expression analysis of FOXP3, tight junction protein 1 (TJP1), gap junction (GJA1), IDO and CD103 genes was evaluated by real-time PCR. RESULTS The ZO level was higher in CD patients compared to subjects with a normal small bowel mucosa, particularly in those with Marsh IIIc atrophy (p = 0.01), and correlated with the density of EV (r = 0.63; p = 0.0003) and IDO+ DCs (r = 0.58; p = 0.01) in the small bowel mucosa. The density of GAD65+ epithelial cells was correlated with the density of EV (r = 0.59; p = 0.03) and IDO+ DCs (r = 0.78; p = 0.004) in CD patients. The relative expression of FOXP3 mRNA in the small bowel mucosa tissue was significantly higher in patients with CD, compared to subjects with a normal mucosa, and correlated with the density of EV (r = 0.62; p = 0.017) as well as with the relative expression of IDO mRNA (r = 0.54; p = 0.019). CONCLUSIONS The CD is associated with elevation of the circulating ZO level, the value of which correlates with the density of EV in CD patients with severe atrophic changes in the small bowel mucosa, particularly in cases of concomitant T1D. The CD is also characterized by the close relationship of the density of GAD65+ epithelial cells with the EV, ZO level and IDO+ DCs.
Collapse
MESH Headings
- Adolescent
- Antibodies, Viral/immunology
- Antigens, CD/genetics
- Autoantibodies/immunology
- Case-Control Studies
- Celiac Disease/complications
- Celiac Disease/metabolism
- Celiac Disease/pathology
- Celiac Disease/virology
- Child
- Child, Preschool
- Cholera Toxin/blood
- Connexin 43/genetics
- Dendritic Cells/metabolism
- Dendritic Cells/pathology
- Diabetes Mellitus, Type 1/complications
- Enterovirus/immunology
- Enzyme-Linked Immunosorbent Assay
- Female
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Glutamate Decarboxylase/immunology
- Glutamate Decarboxylase/metabolism
- Haptoglobins
- Humans
- Immunoglobulin A/immunology
- Immunohistochemistry
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Integrin alpha Chains/genetics
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Intestinal Mucosa/virology
- Intestine, Small/metabolism
- Intestine, Small/pathology
- Intestine, Small/virology
- Male
- Permeability
- Protein Precursors
- RNA, Messenger/metabolism
- Real-Time Polymerase Chain Reaction
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
- Zonula Occludens-1 Protein/genetics
Collapse
Affiliation(s)
- Tamara Vorobjova
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 51014, Tartu, Estonia.
| | - Helerin Raikkerus
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 51014, Tartu, Estonia
| | - Lumme Kadaja
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Ija Talja
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 51014, Tartu, Estonia
| | - Oivi Uibo
- Department of Pediatrics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Children's Clinic, Tartu University Hospital, Tartu, Estonia
| | - Kaire Heilman
- Department of Pediatrics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Tallinn Children's Hospital, Tallinn, Estonia
| | - Raivo Uibo
- Department of Immunology, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 19, 51014, Tartu, Estonia
| |
Collapse
|
121
|
Lundberg M, Seiron P, Ingvast S, Korsgren O, Skog O. Insulitis in human diabetes: a histological evaluation of donor pancreases. Diabetologia 2017; 60:346-353. [PMID: 27796420 PMCID: PMC6518093 DOI: 10.1007/s00125-016-4140-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/27/2016] [Indexed: 01/05/2023]
Abstract
AIMS/HYPOTHESIS According to the consensus criteria developed for type 1 diabetes, an individual can be diagnosed with insulitis when ≥ 15 CD45+ cells are found within the parenchyma or in the islet-exocrine interface in ≥ 3 islets. The aim of this study was to determine the frequency of individuals with type 2 diabetes fulfilling these criteria with reference to non-diabetic and type 1 diabetic individuals. METHODS Insulitis was determined by examining CD45+ cells in the pancreases of 50, 13 and 44 organ donors with type 2 diabetes, type 1 diabetes and no diabetes, respectively. CD3+ cells (T cells) infiltrating the islets were evaluated in insulitic donors. In insulitic donors with type 2 diabetes, the pancreases were characterised according to the presence of CD68 (macrophages), myeloperoxidase (MPO; neutrophils), CD3, CD20 (B cells) and HLA class I hyperstained islets. In all type 2 diabetic donors, potential correlations of insulitis with dynamic glucose-stimulated insulin secretion in vitro or age, BMI, HbA1c or autoantibody positivity were examined. RESULTS Overall, 28% of the type 2 diabetic donors fulfilled the consensus criteria for insulitis developed for type 1 diabetes. Of the type 1 diabetic donors, 31% fulfilled the criteria. None of the non-diabetic donors met the criteria. Only type 1 diabetic donors had ≥ 15 CD3+ cells in ≥ 3 islets. Type 2 diabetic donors with insulitis also had a substantial number of CD45+ cells in the exocrine parenchyma. Macrophages constituted the largest fraction of CD45+ cells, followed by neutrophils and T cells. Of type 2 diabetic pancreases with insulitis, 36% contained islets that hyperstained for HLA class I. Isolated islets from type 2 diabetic donors secreted less insulin than controls, although with preserved dynamics. Insulitis in the type 2 diabetic donors did not correlate with glucose-stimulated insulin secretion, the presence of autoantibodies, BMI or HbA1c. CONCLUSIONS/INTERPRETATION The current definition of insulitis cannot be used to distinguish pancreases retrieved from individuals with type 1 diabetes from those with type 2 diabetes. On the basis of our findings, we propose a revised definition of insulitis, with a positive diagnosis when ≥ 15 CD3+ cells, not CD45+ cells, are found in ≥ 3 islets.
Collapse
Affiliation(s)
- Marcus Lundberg
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory C11, Clinical Immunology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden.
| | - Peter Seiron
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory C11, Clinical Immunology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden
| | - Sofie Ingvast
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory C11, Clinical Immunology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory C11, Clinical Immunology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden
| | - Oskar Skog
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory C11, Clinical Immunology, Uppsala University, Dag Hammarskjölds väg 20, 751 85, Uppsala, Sweden
| |
Collapse
|
122
|
Echovirus 6 Infects Human Exocrine and Endocrine Pancreatic Cells and Induces Pro-Inflammatory Innate Immune Response. Viruses 2017; 9:v9020025. [PMID: 28146100 PMCID: PMC5332944 DOI: 10.3390/v9020025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/16/2017] [Indexed: 12/22/2022] Open
Abstract
Human enteroviruses (HEV), especially coxsackievirus serotype B (CVB) and echovirus (E), have been associated with diseases of both the exocrine and endocrine pancreas, but so far evidence on HEV infection in human pancreas has been reported only in islets and ductal cells. This study aimed to investigate the capability of echovirus strains to infect human exocrine and endocrine pancreatic cells. Infection of explanted human islets and exocrine cells with seven field strains of E6 caused cytopathic effect, virus titer increase and production of HEV protein VP1 in both cell types. Virus particles were found in islets and acinar cells infected with E6. No cytopathic effect or infectious progeny production was observed in exocrine cells exposed to the beta cell-tropic strains of E16 and E30. Endocrine cells responded to E6, E16 and E30 by upregulating the transcription of interferon-induced with helicase C domain 1 (IF1H1), 2′-5′-oligoadenylate synthetase 1 (OAS1), interferon-β (IFN-β), chemokine (C–X–C motif) ligand 10 (CXCL10) and chemokine (C–C motif) ligand 5 (CCL5). Echovirus 6, but not E16 or E30, led to increased transcription of these genes in exocrine cells. These data demonstrate for the first time that human exocrine cells represent a target for E6 infection and suggest that certain HEV serotypes can replicate in human pancreatic exocrine cells, while the pancreatic endocrine cells are permissive to a wider range of HEV.
Collapse
|
123
|
Alidjinou EK, Engelmann I, Bossu J, Villenet C, Figeac M, Romond MB, Sané F, Hober D. Persistence of Coxsackievirus B4 in pancreatic ductal-like cells results in cellular and viral changes. Virulence 2017; 8:1229-1244. [PMID: 28112573 DOI: 10.1080/21505594.2017.1284735] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Although known as cytolytic viruses, group B coxackieviruses (CVB) are able to establish a persistent infection in vitro and in vivo. Viral persistence has been reported as a key mechanism in the pathogenesis of CVB-associated chronic diseases such as type 1 diabetes (T1D). The impact of CVB4 persistence on human pancreas ductal-like cells was investigated. METHODS A persistent CVB4 infection was established in ductal-like cells. PDX-1 expression, resistance to CVB4-induced lysis and CAR expression were evaluated. The profile of cellular microRNAs (miRNAs) was investigated through miRNA-sequencing. Viral phenotypic changes were examined, and genomic modifications were assessed by sequencing of the viral genome. RESULTS The CVB4 persistence in ductal-like cells was productive, with continuous release of infectious particles. Persistently infected cells displayed a resistance to CVB4-induced lysis upon superinfection and expression of PDX-1 and CAR was decreased. These changes were maintained even after virus clearance. The patterns of cellular miRNA expression in mock-infected and in CVB4-persistently infected ductal-like cells were clearly different. The persistent infection-derived virus (PIDV) was still able to induce cytopathic effect but its plaques were smaller than the parental virus. Several mutations appeared in various PIDV genome regions, but amino acid substitutions did not affect the predicted site of interaction with CAR. CONCLUSION Cellular and viral changes occur during persistent infection of human pancreas ductal-like cells with CVB4. The persistence of cellular changes even after virus clearance supports the hypothesis of a long-lasting impact of persistent CVB infection on the cells.
Collapse
Affiliation(s)
- E K Alidjinou
- a Univ Lille, CHU Lille, Laboratoire de Virologie EA3610 , Lille , France
| | - I Engelmann
- a Univ Lille, CHU Lille, Laboratoire de Virologie EA3610 , Lille , France
| | - J Bossu
- a Univ Lille, CHU Lille, Laboratoire de Virologie EA3610 , Lille , France
| | - C Villenet
- b Plate-forme de Génomique Fonctionnelle et Structurale , CHU de Lille , France
| | - M Figeac
- b Plate-forme de Génomique Fonctionnelle et Structurale , CHU de Lille , France
| | - M-B Romond
- a Univ Lille, CHU Lille, Laboratoire de Virologie EA3610 , Lille , France
| | - F Sané
- a Univ Lille, CHU Lille, Laboratoire de Virologie EA3610 , Lille , France
| | - D Hober
- a Univ Lille, CHU Lille, Laboratoire de Virologie EA3610 , Lille , France
| |
Collapse
|
124
|
Bayer AL, Fraker CA. The Folate Cycle As a Cause of Natural Killer Cell Dysfunction and Viral Etiology in Type 1 Diabetes. Front Endocrinol (Lausanne) 2017; 8:315. [PMID: 29218028 PMCID: PMC5703744 DOI: 10.3389/fendo.2017.00315] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/30/2017] [Indexed: 12/15/2022] Open
Abstract
The folate pathway is critical to proper cellular function and metabolism. It is responsible for multiple functions, including energy (ATP) production, methylation reactions for DNA and protein synthesis and the production of immunomodulatory molecules, inosine and adenosine. These play an important role in immune signaling and cytotoxicity. Herein, we hypothesize that defects in the folate pathway in genetically susceptible individuals could lead to immune dysfunction, permissive environments for chronic cyclical latent/lytic viral infection, and, ultimately, the development of unchecked autoimmune responses to infected tissue, in this case islet beta cells. In the context of type 1 diabetes (T1D), there has been a recent increase in newly diagnosed cases of T1D in the past 20 years that has exceeded previous epidemiological predictions with yet unidentified factor(s). This speaks to a potential environmental trigger that adversely affects immune responses. Most research into the immune dysfunction of T1D has focused on downstream adaptive responses of T and B cells neglecting the role of the upstream innate players such as natural killer (NK) cells. Constantly, surveilling the blood and tissues for pathogens, NK cells remove threats through direct cytotoxic responses and recruitment of adaptive responses using cytokines, such as IL-1β and IFN-γ. One long-standing hypothesis suggests viral infection as a potential trigger for the autoimmune response in T1D. Recent data suggest multiple viruses as potential causal agents. Intertwined with this is an observed reduced NK cell enumeration, cytotoxicity, and cytokine signaling in T1D patients. Many of the viruses implicated in T1D are chronic latent/lysogenic infections with demonstrated capacity to reduce NK cell response and number through mechanisms that resemble those of pregnancy tolerance. Defects in the folate pathway in T1D patients could result in decreased immune response to viral infection or viral reactivation. Dampened NK responses to infections result in improper signaling, improper antigen presentation, and amplified CD8+ lymphocyte proliferation and cytotoxicity, a hallmark of beta cell infiltrates in patients with T1D onset. This would suggest a critical role for NK cells in T1D development linked to viral infection and the importance of the folate pathway in maintaining proper NK response.
Collapse
Affiliation(s)
- Allison L. Bayer
- Immunobiology Laboratory, Leonard M. Miller School of Medicine, Diabetes Research Institute, University of Miami, Miami, FL, United States
| | - Christopher A. Fraker
- Tissue and Biomedical Engineering Laboratory, Leonard M. Miller School of Medicine, Diabetes Research Institute, University of Miami, Miami, FL, United States
- *Correspondence: Christopher A. Fraker,
| |
Collapse
|
125
|
Domsgen E, Lind K, Kong L, Hühn MH, Rasool O, van Kuppeveld F, Korsgren O, Lahesmaa R, Flodström-Tullberg M. An IFIH1 gene polymorphism associated with risk for autoimmunity regulates canonical antiviral defence pathways in Coxsackievirus infected human pancreatic islets. Sci Rep 2016; 6:39378. [PMID: 28000722 PMCID: PMC5175199 DOI: 10.1038/srep39378] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 11/23/2016] [Indexed: 02/08/2023] Open
Abstract
The IFIH1 gene encodes the pattern recognition receptor MDA5. A common polymorphism in IFIH1 (rs1990760, A946T) confers increased risk for autoimmune disease, including type 1-diabetes (T1D). Coxsackievirus infections are linked to T1D and cause beta-cell damage in vitro. Here we demonstrate that the rs1990760 polymorphism regulates the interferon (IFN) signature expressed by human pancreatic islets following Coxsackievirus infection. A strong IFN signature was associated with high expression of IFNλ1 and IFNλ2, linking rs1990760 to the expression of type III IFNs. In the high-responding genotype, IRF-1 expression correlated with that of type III IFN, suggesting a positive-feedback on type III IFN transcription. In summary, our study uncovers an influence of rs1990760 on the canonical effector function of MDA5 in response to an acute infection of primary human parenchymal cells with a clinically relevant virus linked to human T1D. It also highlights a previously unrecognized connection between the rs1990760 polymorphism and the expression level of type III IFNs.
Collapse
Affiliation(s)
- Erna Domsgen
- The Center for Infectious Medicine, Department of Medicine HS, Karolinska Institutet, Karolinska University Hospital, Stockholm, 141 86, Sweden
| | - Katharina Lind
- The Center for Infectious Medicine, Department of Medicine HS, Karolinska Institutet, Karolinska University Hospital, Stockholm, 141 86, Sweden
| | - Lingjia Kong
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, 205 20, Finland
| | - Michael H Hühn
- The Center for Infectious Medicine, Department of Medicine HS, Karolinska Institutet, Karolinska University Hospital, Stockholm, 141 86, Sweden
| | - Omid Rasool
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, 205 20, Finland
| | - Frank van Kuppeveld
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584, The Netherlands
| | - Olle Korsgren
- Department of Immunology, Genetics, and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, 751 05, Sweden
| | - Riitta Lahesmaa
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, 205 20, Finland
| | - Malin Flodström-Tullberg
- The Center for Infectious Medicine, Department of Medicine HS, Karolinska Institutet, Karolinska University Hospital, Stockholm, 141 86, Sweden.,Institute of Biosciences and Medical Technologies, University of Tampere, Tampere, 33520, Finland
| |
Collapse
|
126
|
Isaacs SR, Wang J, Kim KW, Yin C, Zhou L, Mi QS, Craig ME. MicroRNAs in Type 1 Diabetes: Complex Interregulation of the Immune System, β Cell Function and Viral Infections. Curr Diab Rep 2016; 16:133. [PMID: 27844276 DOI: 10.1007/s11892-016-0819-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Since the discovery of the first mammalian microRNA (miRNA) more than two decades ago, a plethora of miRNAs has been identified in humans, now amounting to more than 2500. Essential for post-transcriptional regulation of gene networks integral for developmental pathways and immune response, it is not surprising that dysregulation of miRNAs is often associated with the aetiology of complex diseases including cancer, diabetes and autoimmune disorders. Despite massive expansion of small RNA studies and extensive investigation in diverse disease contexts, the role of miRNAs in type 1 diabetes has only recently been explored. Key studies using human islets have recently implicated virus-induced miRNA dysregulation as a pivotal mechanism of β cell destruction, while the interplay between miRNAs, the immune system and β cell survival has been illustrated in studies using animal and cellular models of disease. The role of specific miRNAs as major players in immune system homeostasis highlights their exciting potential as therapeutics and prognostic biomarkers of type 1 diabetes.
Collapse
Affiliation(s)
- Sonia R Isaacs
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- UNSW and POWH Virology Research Laboratory, Prince of Wales Hospital, Sydney, NSW, 2031, Australia
| | - Jie Wang
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Dermatology, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Ki Wook Kim
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia
- UNSW and POWH Virology Research Laboratory, Prince of Wales Hospital, Sydney, NSW, 2031, Australia
| | - Congcong Yin
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Dermatology, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Li Zhou
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Dermatology, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Internal Medicine, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Qing Sheng Mi
- Henry Ford Immunology Program, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Dermatology, Henry Ford Health System, Detroit, MI, 48202, USA
- Department of Internal Medicine, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Maria E Craig
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia.
- UNSW and POWH Virology Research Laboratory, Prince of Wales Hospital, Sydney, NSW, 2031, Australia.
- Institute of Endocrinology and Diabetes, The Children's Hospital at Westmead, Sydney, NSW, 2145, Australia.
- Discipline of Child and Adolescent Health, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
127
|
Abstract
Treatments for autoimmune diseases including type 1 diabetes (T1D) are aimed at resetting the immune system, especially its adaptive arm. The innate immune system is often ignored in the design of novel immune-based therapies. There is increasing evidence for multiple natural killer (NK) subpopulations, but their role is poorly understood in autoimmunity and likely is contributing to the controversial role reported for NKs. In this review, we will summarize NK subsets and their roles in tolerance, autoimmune diabetes, and immunotherapy.
Collapse
Affiliation(s)
- Chris Fraker
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Allison L Bayer
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
128
|
Ettischer-Schmid N, Normann A, Sauter M, Kraft L, Kalbacher H, Kandolf R, Flehmig B, Klingel K. A new monoclonal antibody (Cox mAB 31A2) detects VP1 protein of coxsackievirus B3 with high sensitivity and specificity. Virchows Arch 2016; 469:553-562. [PMID: 27566306 DOI: 10.1007/s00428-016-2008-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/14/2016] [Accepted: 08/18/2016] [Indexed: 12/24/2022]
Abstract
Human enteroviruses, e.g. coxsackieviruses, induce a variety of severe acute and chronic forms of disease, including myocarditis, meningitis and diabetes mellitus type 1. To visualize enterovirus infection with a diagnostic intent, many studies have applied a commercially available antibody (anti-CVB5 VP1, clone 5-D8/1, Dako, Hamburg, Germany) that identifies VP1 of different enteroviral serotypes. Many antibodies, however, have been found to bind non-specifically to proteins of cardiomyocytes and in the interstitial space, resulting in non-specific staining in immunohistochemistry. In this paper we show that the anti-CVB5 VP1 antibody, recognizing VP1 of coxsackieviruses and widely used in diagnostics and research, shows strong cross-reactivity with cellular proteins in the heart (and pancreas) of humans and mice, which calls for a more specific antibody to be used for diagnostic purposes. We observed by Western blot analyses of lysates from human heart tissue samples and HeLa cells two cross-reactive bands when using clone 5-D8/1. Peptide mass fingerprinting (MALDI-TOF) identified these proteins as creatine kinase (B-type) and tubulin, confirming that this mAb detects cellular proteins in addition to viral VP1. In order to overcome the problems of false positive VP1 staining we generated a new highly specific and sensitive monoclonal antibody (Cox mAB 31A2) that recognizes VP1 from CVB3. The new antibody was characterized and was found to function well in immunohistochemistry, immunofluorescence staining, Western blotting, ELISA and FACS analyses.
Collapse
Affiliation(s)
- Nicole Ettischer-Schmid
- Institute for Pathology, Department of Molecular Pathology, University Hospital of Tuebingen, Liebermeisterstrasse 8, D-72076, Tuebingen, Germany
| | | | - Martina Sauter
- Institute for Pathology, Department of Molecular Pathology, University Hospital of Tuebingen, Liebermeisterstrasse 8, D-72076, Tuebingen, Germany
| | - Lisa Kraft
- Institute for Pathology, Department of Molecular Pathology, University Hospital of Tuebingen, Liebermeisterstrasse 8, D-72076, Tuebingen, Germany
- Interfaculty Institute of Biochemistry, University of Tuebingen, D-72076, Tuebingen, Germany
| | - Hubert Kalbacher
- Interfaculty Institute of Biochemistry, University of Tuebingen, D-72076, Tuebingen, Germany
| | - Reinhard Kandolf
- Institute for Pathology, Department of Molecular Pathology, University Hospital of Tuebingen, Liebermeisterstrasse 8, D-72076, Tuebingen, Germany
| | | | - Karin Klingel
- Institute for Pathology, Department of Molecular Pathology, University Hospital of Tuebingen, Liebermeisterstrasse 8, D-72076, Tuebingen, Germany.
| |
Collapse
|
129
|
Rodriguez-Calvo T, Sabouri S, Anquetil F, von Herrath MG. The viral paradigm in type 1 diabetes: Who are the main suspects? Autoimmun Rev 2016; 15:964-9. [PMID: 27491567 DOI: 10.1016/j.autrev.2016.07.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 07/08/2016] [Indexed: 12/23/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by the loss of pancreatic beta cells in the islets of Langerhans. Although genetic predisposition plays an important role in T1D development, studies of identical twins suggest that environmental factors such as viruses and other pathogens may be critical triggers either through direct cytolytic effect and gradual beta cell destruction, or by bystander activation of the immune system. In addition, viruses may circumvent the host immune response and have the capacity to establish chronic lifelong infections. The association of various viral infections with the induction of T1D has been extensively studied at the serological and epidemiological level. However, there is still little evidence from studies of human pancreas to confirm their presence or a causal role in disease pathogenesis. In this review, we identify possible suspects for viral triggers of disease and explain their potential roles in the "viral paradigm" of T1D.
Collapse
Affiliation(s)
- Teresa Rodriguez-Calvo
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Somayeh Sabouri
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Florence Anquetil
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Matthias G von Herrath
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA; Novo Nordisk Diabetes Research & Development Center, Seattle, WA, USA.
| |
Collapse
|
130
|
Abstract
Environmental factors play an important role in the pathogenesis of type 1 diabetes and can determine if a genetically susceptible individual develops the disease. Increasing evidence suggest that among other exogenous agents certain virus infections can contribute to the beta-cell damaging process. Possible viral etiology of type 1 diabetes has been explored extensively but the final proof for causality is still lacking. Currently, the group of enteroviruses (EVs) is considered as the strongest candidate. These viruses have been found in the pancreas of type 1 diabetic patients, and epidemiological studies have shown more EV infections in diabetic patients than in controls. Prospective studies, such as the Type 1 Diabetes Prediction and Prevention (DIPP) study in Finland, are of fundamental importance in the evaluation viral effects as they can cover all stages of the beta-cell damaging process, including those preceding the initiation of the process. DIPP study has carried out the most comprehensive virological analyses ever done in prospective cohorts. This article summarizes the findings from these analyses and discuss them in the context of the existing other knowledge and the prospects for intervention studies with EV vaccines or antiviral drugs.
Collapse
Affiliation(s)
- Heikki Hyöty
- Department of Virology, School of Medicine, University of Tampere, Tampere, Finland.,Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| |
Collapse
|
131
|
Abstract
The incidence of type 1 diabetes has risen considerably in the past 30 years due to changes in the environment that have been only partially identified. In this Series paper, we critically discuss candidate triggers of islet autoimmunity and factors thought to promote progression from autoimmunity to overt type 1 diabetes. We revisit previously proposed hypotheses to explain the growth in the incidence of type 1 diabetes in light of current data. Finally, we suggest a unified model in which immune tolerance to β cells can be broken by several environmental exposures that induce generation of hybrid peptides acting as neoautoantigens.
Collapse
Affiliation(s)
- Marian Rewers
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Johnny Ludvigsson
- Division of Pediatrics, Department of Clinical and Experimental Medicine, Medical Faculty, Linköping University and Linköping University Hospital, Linköping, Sweden.
| |
Collapse
|
132
|
Abstract
Type 1 diabetes mellitus (T1DM) is caused by progressive autoimmune-mediated loss of pancreatic β-cell mass via apoptosis. The onset of T1DM depends on environmental factors that interact with predisposing genes to induce an autoimmune assault against β cells. Epidemiological, clinical and pathology studies in humans support viral infection--particularly by enteroviruses (for example, coxsackievirus)--as an environmental trigger for the development of T1DM. Many candidate genes for T1DM, such as MDA5, PTPN2 and TYK2, regulate antiviral responses in both β cells and the immune system. Cellular permissiveness to viral infection is modulated by innate antiviral responses that vary among different tissues or cell types. Some data indicate that pancreatic islet α cells trigger a more efficient antiviral response to infection with diabetogenic viruses than do β cells, and so are able to eradicate viral infections without undergoing apoptosis. This difference could account for the varying ability of islet-cell subtypes to clear viral infections and explain why chronically infected pancreatic β cells, but not α cells, are targeted by an autoimmune response and killed during the development of T1DM. These issues and attempts to target viral infection as a preventive therapy for T1DM are discussed in the present Review.
Collapse
Affiliation(s)
- Anne Op de Beeck
- Center for Diabetes Research, Universite Libre de Bruxelles, 808 Route de Lennik, CP618, B-1070, Brussels, Belgium
| | - Decio L Eizirik
- Center for Diabetes Research, Universite Libre de Bruxelles, 808 Route de Lennik, CP618, B-1070, Brussels, Belgium
- Welbio, Universite Libre de Bruxelles, 808 Route de Lennik, CP618, B-1070, Brussels, Belgium
| |
Collapse
|
133
|
Cabrera SM, Henschel AM, Hessner MJ. Innate inflammation in type 1 diabetes. Transl Res 2016; 167:214-27. [PMID: 25980926 PMCID: PMC4626442 DOI: 10.1016/j.trsl.2015.04.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/02/2015] [Accepted: 04/21/2015] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes mellitus (T1D) is an autoimmune disease often diagnosed in childhood that results in pancreatic β-cell destruction and life-long insulin dependence. T1D susceptibility involves a complex interplay between genetic and environmental factors and has historically been attributed to adaptive immunity, although there is now increasing evidence for a role of innate inflammation. Here, we review studies that define a heightened age-dependent innate inflammatory state in T1D families that is paralleled with high fidelity by the T1D-susceptible biobreeding rat. Innate inflammation may be driven by changes in interactions between the host and environment, such as through an altered microbiome, intestinal hyperpermeability, or viral exposures. Special focus is put on the temporal measurement of plasma-induced transcriptional signatures of recent-onset T1D patients and their siblings as well as in the biobreeding rat as it defines the natural history of innate inflammation. These sensitive and comprehensive analyses have also revealed that those who successfully managed T1D risk develop an age-dependent immunoregulatory state, providing a possible mechanism for the juvenile nature of T1D. Therapeutic targeting of innate inflammation has been proven effective in preventing and delaying T1D in rat models. Clinical trials of agents that suppress innate inflammation have had more modest success, but efficacy may be improved by the addition of combinatorial approaches that target other aspects of T1D pathogenesis. An understanding of innate inflammation and mechanisms by which this susceptibility is both potentiated and mitigated offers important insight into T1D progression and avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Susanne M. Cabrera
- The Max McGee National Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, and Department of Pediatrics at the Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Angela M. Henschel
- The Max McGee National Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, and Department of Pediatrics at the Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Martin J. Hessner
- The Max McGee National Research Center for Juvenile Diabetes, Children’s Research Institute of Children’s Hospital of Wisconsin, and Department of Pediatrics at the Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
134
|
Hodik M, Skog O, Lukinius A, Isaza-Correa JM, Kuipers J, Giepmans BNG, Frisk G. Enterovirus infection of human islets of Langerhans affects β-cell function resulting in disintegrated islets, decreased glucose stimulated insulin secretion and loss of Golgi structure. BMJ Open Diabetes Res Care 2016; 4:e000179. [PMID: 27547409 PMCID: PMC4985798 DOI: 10.1136/bmjdrc-2015-000179] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 04/27/2016] [Accepted: 05/02/2016] [Indexed: 12/16/2022] Open
Abstract
AIMS/HYPOTHESIS In type 1 diabetes (T1D), most insulin-producing β cells are destroyed, but the trigger is unknown. One of the possible triggers is a virus infection and the aim of this study was to test if enterovirus infection affects glucose stimulated insulin secretion and the effect of virus replication on cellular macromolecules and organelles involved in insulin secretion. METHODS Isolated human islets were infected with different strains of coxsackievirus B (CVB) virus and the glucose-stimulated insulin release (GSIS) was measured in a dynamic perifusion system. Classical morphological electron microscopy, large-scale electron microscopy, so-called nanotomy, and immunohistochemistry were used to study to what extent virus-infected β cells contained insulin, and real-time PCR was used to analyze virus induced changes of islet specific genes. RESULTS In islets infected with CVB, GSIS was reduced in correlation with the degree of virus-induced islet disintegration. The expression of the gene encoding insulin was decreased in infected islets, whereas the expression of glucagon was not affected. Also, in islets that were somewhat disintegrated, there were uninfected β cells. Ultrastructural analysis revealed that virus particles and virus replication complexes were only present in β cells. There was a significant number of insulin granules remaining in the virus-infected β cells, despite decreased expression of insulin mRNA. In addition, no typical Golgi apparatus was detected in these cells. Exposure of islets to synthetic dsRNA potentiated glucose-stimulated insulin secretion. CONCLUSIONS/INTERPRETATION Glucose-stimulated insulin secretion; organelles involved in insulin secretion and gene expression were all affected by CVB replication in β cells.
Collapse
Affiliation(s)
- M Hodik
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - O Skog
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - A Lukinius
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - J M Isaza-Correa
- Department of Cell Biology, University Medical Center Groningen, University Groningen, Groningen, The Netherlands
| | - J Kuipers
- Department of Cell Biology, University Medical Center Groningen, University Groningen, Groningen, The Netherlands
| | - B N G Giepmans
- Department of Cell Biology, University Medical Center Groningen, University Groningen, Groningen, The Netherlands
| | - G Frisk
- Department of Immunology, Genetics and Pathology, The Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
135
|
Smura T, Natri O, Ylipaasto P, Hellman M, Al-Hello H, Piemonti L, Roivainen M. Enterovirus strain and type-specific differences in growth kinetics and virus-induced cell destruction in human pancreatic duct epithelial HPDE cells. Virus Res 2015; 210:188-97. [DOI: 10.1016/j.virusres.2015.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/31/2015] [Accepted: 08/05/2015] [Indexed: 12/16/2022]
|
136
|
Rodriguez-Calvo T, Suwandi JS, Amirian N, Zapardiel-Gonzalo J, Anquetil F, Sabouri S, von Herrath MG. Heterogeneity and Lobularity of Pancreatic Pathology in Type 1 Diabetes during the Prediabetic Phase. J Histochem Cytochem 2015. [PMID: 26216138 DOI: 10.1369/0022155415576543] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which insulin-producing beta cells are destroyed in the islets of Langerhans. One of its main pathological manifestations is the hyper-expression of Major Histocompatibility Complex I (MHC-I) by beta cells, which was first described over 3 decades ago yet its cause remains unknown. It might not only be a sign of beta cell dysfunction but could also render the cells susceptible to autoimmune destruction; for example, by islet-infiltrating CD8 T cells. In this report, we studied pancreas tissue from a 22-year-old non-diabetic male cadaveric organ donor who had been at high risk of developing T1D, in which autoantibodies against GAD and IA-2 were detected. Pancreas sections were analyzed for signs of inflammation. Multiple insulin-containing islets were identified, which hyper-expressed MHC-I. However, islet density and MHC-I expression exhibited a highly lobular and heterogeneous pattern even within the same section. In addition, many islets with high expression of MHC-I presented higher levels of CD8 T cell infiltration than normal islets. These results demonstrate the heterogeneity of human pathology that occurs early during the pre-diabetic, autoantibody positive phase, and should contribute to the understanding of human T1D.
Collapse
Affiliation(s)
- Teresa Rodriguez-Calvo
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California (TRC, JSS, NA, JZG, FA, SS, MGVH)
| | - Jessica S Suwandi
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California (TRC, JSS, NA, JZG, FA, SS, MGVH),Department of Immunohematology & Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands (JSS)
| | - Natalie Amirian
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California (TRC, JSS, NA, JZG, FA, SS, MGVH)
| | - Jose Zapardiel-Gonzalo
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California (TRC, JSS, NA, JZG, FA, SS, MGVH)
| | - Florence Anquetil
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California (TRC, JSS, NA, JZG, FA, SS, MGVH)
| | - Somayeh Sabouri
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California (TRC, JSS, NA, JZG, FA, SS, MGVH)
| | - Matthias G von Herrath
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California (TRC, JSS, NA, JZG, FA, SS, MGVH),Novo Nordisk Diabetes Research & Development Center, Seattle, Washington (MGVH)
| |
Collapse
|
137
|
Abstract
Type 1 diabetes (T1D) results from genetic predisposition and environmental factors leading to the autoimmune destruction of pancreatic beta cells. Recently, a rapid increase in the incidence of childhood T1D has been observed worldwide; this is too fast to be explained by genetic factors alone, pointing to the spreading of environmental factors linked to the disease. Enteroviruses (EVs) are perhaps the most investigated environmental agents in relationship to the pathogenesis of T1D. While several studies point to the likelihood of such correlation, epidemiological evidence in its support is inconclusive or in some instances even against it. Hence, it is still unknown if and how EVs are involved in the development of T1D. Here we review recent findings concerning the biology of EV in beta cells and the potential implications of this knowledge for the understanding of beta cell dysfunction and autoimmune destruction in T1D.
Collapse
Affiliation(s)
- Antje Petzold
- />Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Fetscherstr.74, 01307 Dresden, Germany
- />German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Michele Solimena
- />Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Fetscherstr.74, 01307 Dresden, Germany
- />German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- />Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Klaus-Peter Knoch
- />Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Fetscherstr.74, 01307 Dresden, Germany
- />German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| |
Collapse
|
138
|
Alidjinou EK, Chehadeh W, Weill J, Vantyghem MC, Stuckens C, Decoster A, Hober C, Hober D. Monocytes of Patients with Type 1 Diabetes Harbour Enterovirus RNA. Eur J Clin Invest 2015; 45:918-24. [PMID: 26108863 DOI: 10.1111/eci.12485] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 06/20/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Intracellular enterovirus (EV) RNA was detected in blood of patients with type 1 diabetes (T1D). The presence of EV RNA in subsets of peripheral blood mononuclear cells (PBMCs) of patients, and the in vitro infection of these cells with an EV, was investigated. MATERIALS AND METHODS Blood was collected from 42 patients with T1D, PBMCs were isolated and monocytes were purified. Interferon alpha (IFNα) mRNA and EV RNA were investigated using RT-PCR. Levels of IFNα in plasma were measured using an immunoassay. Cells were inoculated with Coxsackievirus B4 (CBV4) in vitro, and infection was assessed by indirect immunofluorescence (IFI). RESULTS Interferon alpha mRNA was detected in blood and in monocytes of 12 of 42 patients with T1D, but not in monocyte-depleted PBMCs of the same individuals. Significant plasma levels of IFNα (≥ 5 IU/mL) were found in six patients. EV RNA was detected in whole blood and in monocytes of seven patients and negative-strand EV RNA was found in monocytes of 6 of them. When monocytes of patients with IFNα and/or EV RNA in their blood were inoculated with CVB4, the proportion of cells stained by an anti-VP1 antibody was 8.8 ± 1%, whereas no VP1 was detected in the monocytes of IFNα, EV RNA negative patients. Nevertheless, when CBV4 was mixed with plasma, VP1 was detected in monocytes of all patients with T1D (staining ranging from 12 to 36%). CONCLUSIONS Our data indicate that monocytes of patients with T1D can harbor EV RNA and IFNα mRNA and can be infected with an EV in vitro.
Collapse
Affiliation(s)
| | - Wassim Chehadeh
- Laboratoire de virologie EA3610, Université de Lille, CHU Lille, France
| | - Jacques Weill
- Unité d'Endocrinologie pédiatrique, CHU Lille, France
| | | | | | - Anne Decoster
- Laboratoire, Institut Catholique de Lille, Hôpital St Philibert, Lomme, France
| | - Christine Hober
- Service de Diabétologie, Centre Hospitalier, Hénin-Beaumont, France
| | - Didier Hober
- Laboratoire de virologie EA3610, Université de Lille, CHU Lille, France
| |
Collapse
|
139
|
Taniguchi K, Russell MA, Richardson SJ, Morgan NG. The subcellular distribution of cyclin-D1 and cyclin-D3 within human islet cells varies according to the status of the pancreas donor. Diabetologia 2015; 58:2056-63. [PMID: 26055066 DOI: 10.1007/s00125-015-3645-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/15/2015] [Indexed: 12/26/2022]
Abstract
AIMS/HYPOTHESIS In humans, the rate of beta cell proliferation declines rapidly during the postnatal period and remains low throughout adult life. Recent studies suggest that this may reflect the distribution of cell cycle regulators which, unusually, are located in the cytosolic compartment of beta cells in islets isolated from adults. In the present work, we examined whether the localisation of cyclin-D molecules is also cytosolic in the islet cells of pancreatic samples studied in situ. METHODS Immunohistochemical approaches were employed to examine the subcellular localisation of cyclin-D1, -D2 and -D3 in human pancreatic samples recovered either from heart-beating donors or post mortem. Immunofluorescence methods were used to reveal the cellular localisation of cyclin-D1 and -D3. RESULTS The distribution of cyclin-D2 was invariably cytosolic in islet cells, whereas the localisation of cyclin-D1 and -D3 varied according to the status of the donor. In pancreatic sections from heart-beating donors these molecules were primarily nuclear. By contrast, in samples collected post mortem, they were mainly cytosolic. Cyclin-D1 was detected only in beta cells whereas cyclin-D3 was detected in both alpha and beta cells. The proportion of donors who were immunopositive for cyclin-D1 declined from 71% in controls to 30% in those with type 1 diabetes. Cyclin-D3 was present in the islets of the majority of donors in both groups. CONCLUSIONS/INTERPRETATION The subcellular localisation of cyclin-D molecules varies according to the status of the donor. Both cyclin-D1 and -D3 can be found in the nuclei of human islet cells in situ.
Collapse
Affiliation(s)
- Kazuto Taniguchi
- Institute of Biomedical & Clinical Science, University of Exeter Medical School, RILD Building, Barrack Road, Exeter, EX2 5DW, UK
| | | | | | | |
Collapse
|
140
|
Heinonen MT, Moulder R, Lahesmaa R. New Insights and Biomarkers for Type 1 Diabetes: Review for Scandinavian Journal of Immunology. Scand J Immunol 2015; 82:244-53. [DOI: 10.1111/sji.12338] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 06/25/2015] [Indexed: 12/16/2022]
Affiliation(s)
- M. T. Heinonen
- Turku Centre for Biotechnology; University of Turku; Åbo Akademi University; Turku Finland
| | - R. Moulder
- Turku Centre for Biotechnology; University of Turku; Åbo Akademi University; Turku Finland
| | - R. Lahesmaa
- Turku Centre for Biotechnology; University of Turku; Åbo Akademi University; Turku Finland
| |
Collapse
|
141
|
Campbell-Thompson M. Organ donor specimens: What can they tell us about type 1 diabetes? Pediatr Diabetes 2015; 16:320-30. [PMID: 25998576 PMCID: PMC4718555 DOI: 10.1111/pedi.12286] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 04/04/2015] [Accepted: 05/01/2015] [Indexed: 12/16/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic disease resulting from the destruction of pancreatic beta cells, due to a poorly understood combination of genetic, environmental, and immune factors. The JDRF Network for Pancreatic Organ donors with Diabetes (nPOD) program recovers transplantation quality pancreas from organ donors throughout the USA. In addition to recovery of donors with T1D, non-diabetic donors include those with islet autoantibodies. Donors with type 2 diabetes and other conditions are also recovered to aid investigations directed at the full spectrum of pathophysiological mechanisms affecting beta cells. One central processing laboratory conducts standardized procedures for sample processing, storage, and distribution, intended for current and future cutting edge investigations. Baseline histology characterizations are performed on the pancreatic samples, with images of the staining results provided though whole-slide digital scans. Uniquely, these high-grade biospecimens are provided without expense to investigators, working worldwide, seeking methods for disease prevention and reversal strategies. Collaborative working groups are highly encouraged, bringing together multiple investigators with different expertise to foster collaborations in several areas of critical need. This mini-review will provide some key histopathological findings emanating from the nPOD collection, including the heterogeneity of beta cell loss and islet inflammation (insulitis), beta cell mass, insulin-producing beta cells in chronic T1D, and pancreas weight reductions at disease onset. Analysis of variations in histopathology observed from these organ donors could provide for mechanistic differences related to etiological agents and serve an important function in terms of identifying the heterogeneity of T1D.
Collapse
Affiliation(s)
- Martha Campbell-Thompson
- The Department of Pathology, Immunology, and Laboratory Medicine, The University of Florida, College of Medicine, Gainesville, FL, USA
| |
Collapse
|
142
|
Marroqui L, Lopes M, dos Santos RS, Grieco FA, Roivainen M, Richardson SJ, Morgan NG, Op de Beeck A, Eizirik DL. Differential cell autonomous responses determine the outcome of coxsackievirus infections in murine pancreatic α and β cells. eLife 2015; 4:e06990. [PMID: 26061776 PMCID: PMC4480275 DOI: 10.7554/elife.06990] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 06/08/2015] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease caused by loss of pancreatic β cells via apoptosis while neighboring α cells are preserved. Viral infections by coxsackieviruses (CVB) may contribute to trigger autoimmunity in T1D. Cellular permissiveness to viral infection is modulated by innate antiviral responses, which vary among different cell types. We presently describe that global gene expression is similar in cytokine-treated and virus-infected human islet cells, with up-regulation of gene networks involved in cell autonomous immune responses. Comparison between the responses of rat pancreatic α and β cells to infection by CVB5 and 4 indicate that α cells trigger a more efficient antiviral response than β cells, including higher basal and induced expression of STAT1-regulated genes, and are thus better able to clear viral infections than β cells. These differences may explain why pancreatic β cells, but not α cells, are targeted by an autoimmune response during T1D. DOI:http://dx.doi.org/10.7554/eLife.06990.001 Type 1 diabetes is caused by a person's immune system attacking the cells in their pancreas that produce insulin. This eventually kills off so many of these cells—known as beta cells—that the pancreas is unable to make enough insulin. As a result, individuals with type 1 diabetes must inject insulin to help their bodies process sugars. One of the mysteries of type 1 diabetes is why the beta cells in the pancreas are killed by the immune system while neighboring alpha cells, which produce the hormone glucagon, are spared. Scientists suspect a combination of genetic and environmental factors contributes to type 1 diabetes. Certain viruses, including one called Coxsackievirus, appear to trigger type 1 diabetes in susceptible individuals. Other factors may also make these individuals more likely to develop the disease. For example, they may ‘express’ genes that are thought to increase the risk of type 1 diabetes, many of which control how the immune system responds to viral infections. These genes may make susceptible individuals experience excessive inflammation, because inflammation is what ultimately kills off the beta cells. Now, Marroqui, Lopes, dos Santos et al. provide evidence that suggests why the alpha cells are spared the immune onslaught in type 1 diabetes. In initial experiments, clusters of cells—known as islets—from the human pancreas were either exposed to small proteins that cause inflammation or infected with the Coxsakievirus. Both events caused a similar increase in the expression of particular immune response genes in the islets. This indicates that these islet cells are able to react to the virus and trigger a first line of defense, which will be further boosted when the immune system is subsequently called into action. Islets contain both alpha and beta cells, and so further experiments on alpha and beta cells from rats investigated whether the two cell types respond differently when infected by the Coxsakievirus. The results revealed that alpha cells boost the expression of the genes needed to clear the virus to a greater extent than the beta cells, and so respond more efficiently to the virus. Therefore, an infection is more likely to establish itself in the beta cells and consequently trigger inflammation and the immune system's attack on the cells. These observations explain one of the puzzling questions in the diabetes field and reinforce the possibility that a long-standing viral infection in beta cells—which seem to have a limited capacity to clear viral infections—may be one of the mechanisms leading to progressive beta cell destruction in type 1 diabetes. This knowledge will help in the search for ways to protect beta cells against both viral infections and the consequent immune assault. DOI:http://dx.doi.org/10.7554/eLife.06990.002
Collapse
Affiliation(s)
- Laura Marroqui
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Miguel Lopes
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Reinaldo S dos Santos
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Fabio A Grieco
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Merja Roivainen
- National Institute for Health and Welfare, Helsinki, Finland
| | - Sarah J Richardson
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Noel G Morgan
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Anne Op de Beeck
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
143
|
van der Pouw Kraan TCTM, Chen WJ, Bunck MCM, van Raalte DH, van der Zijl NJ, van Genugten RE, van Bloemendaal L, Baggen JM, Serné EH, Diamant M, Horrevoets AJG. Metabolic changes in type 2 diabetes are reflected in peripheral blood cells, revealing aberrant cytotoxicity, a viral signature, and hypoxia inducible factor activity. BMC Med Genomics 2015; 8:20. [PMID: 25956355 PMCID: PMC4446948 DOI: 10.1186/s12920-015-0096-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 04/30/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Metabolic syndrome (MetS) is characterized by central obesity, insulin resistance, dysglycemia, and a pro-atherogenic plasma lipid profile. MetS creates a high risk for development of type 2 diabetes (T2DM) and cardiovascular disease (CVD), presumably by altering inflammatory responses. Presently, it is unknown how the chronic metabolic disturbances in acute hyperglycemia, MetS and T2DM affect the immune activity of peripheral blood cells. METHODS We performed genome-wide expression analysis of peripheral blood cells obtained from patients with T2DM (n = 6) and age-, sex- , BMI- and blood pressure-matched obese individuals with MetS (n = 4) and lean healthy normoglycemic controls (n = 3), both under fasting conditions and after controlled induction of acute hyperglycemia during a 70 min hyperglycemic clamp. Differential gene expression during fasting conditions was confirmed by real-time PCR, for which we included additional age-, sex-, BMI-, and blood pressure-matched obese individuals with (n = 4) or without (n = 4) MetS. RESULTS Pathway and Gene ontology analysis applied to baseline expression profiles of peripheral blood cells from MetS and T2DM patients revealed metabolic changes, highly similar to a reoviral infection gene signature in T2DM patients. Transcription factor binding site analysis indicated that increased HIF-1α activity, a transcription factor induced by either hypoxia or oxidative stress, is responsible for this aberrant metabolic profile in peripheral blood cells from T2DM patients. Acute hyperglycemia in healthy controls resulted in reduced expression of cytotoxicity-related genes, representing NK- and CD8(+) cells. In obese controls, MetS and especially T2DM patients, baseline expression of genes involved in cytotoxicity was already low, compared to healthy controls and did not further decrease upon acute hyperglycemia. CONCLUSIONS The reduced activity of cytotoxic genes in T2DM is explained by chronic hyperglycemia, but its acute effects are restricted to healthy controls. Genome expression of circulating leukocytes from T2DM patients differs from MetS individuals by a specific reovirus signature. Our data thus suggest a role for suppressed anti-viral capacity in the etiology of diabetes.
Collapse
Affiliation(s)
| | - Weena J Chen
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Mathijs C M Bunck
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Daniel H van Raalte
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Nynke J van der Zijl
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Renate E van Genugten
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Liselotte van Bloemendaal
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Josefien M Baggen
- Department of Molecular Cell Biology & Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| | - Erik H Serné
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands.
| | - Michaela Diamant
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Anton J G Horrevoets
- Department of Molecular Cell Biology & Immunology, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
144
|
Affiliation(s)
| | - Matthias G von Herrath
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA Novo Nordisk Diabetes Research and Development Center, Seattle, WA
| |
Collapse
|
145
|
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
|
146
|
Gallagher GR, Brehm MA, Finberg RW, Barton BA, Shultz LD, Greiner DL, Bortell R, Wang JP. Viral infection of engrafted human islets leads to diabetes. Diabetes 2015; 64:1358-69. [PMID: 25392246 PMCID: PMC4375078 DOI: 10.2337/db14-1020] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Type 1 diabetes (T1D) is characterized by the destruction of the insulin-producing β-cells of pancreatic islets. Genetic and environmental factors both contribute to T1D development. Viral infection with enteroviruses is a suspected trigger for T1D, but a causal role remains unproven and controversial. Studies in animals are problematic because of species-specific differences in host cell susceptibility and immune responses to candidate viral pathogens such as coxsackievirus B (CVB). In order to resolve the controversial role of viruses in human T1D, we developed a viral infection model in immunodeficient mice bearing human islet grafts. Hyperglycemia was induced in mice by specific ablation of native β-cells. Human islets, which are naturally susceptible to CVB infection, were transplanted to restore normoglycemia. Transplanted mice were infected with CVB4 and monitored for hyperglycemia. Forty-seven percent of CVB4-infected mice developed hyperglycemia. Human islet grafts from infected mice contained viral RNA, expressed viral protein, and had reduced insulin levels compared with grafts from uninfected mice. Human-specific gene expression profiles in grafts from infected mice revealed the induction of multiple interferon-stimulated genes. Thus, human islets can become severely dysfunctional with diminished insulin production after CVB infection of β-cells, resulting in diabetes.
Collapse
Affiliation(s)
- Glen R Gallagher
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Michael A Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Robert W Finberg
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Bruce A Barton
- Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA
| | | | - Dale L Greiner
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Rita Bortell
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Jennifer P Wang
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
147
|
Schulte BM, Gielen PR, Kers-Rebel ED, Schreibelt G, van Kuppeveld FJM, Adema GJ. Enterovirus-infected β-cells induce distinct response patterns in BDCA1+ and BDCA3+ human dendritic cells. PLoS One 2015; 10:e0121670. [PMID: 25806537 PMCID: PMC4373773 DOI: 10.1371/journal.pone.0121670] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 02/03/2015] [Indexed: 12/16/2022] Open
Abstract
Enteroviruses often cause mild disease, yet are also linked to development of autoimmune diabetes. Dendritic cells (DCs) shape both innate and adaptive immune responses, including anti-viral responses. How different human DC subsets shape anti-viral responses, whether they have complementary or overlapping functions and how this relates to autoimmune responses is largely unknown. We used enterovirus-infected β-cells and freshly isolated human myeloid DC (mDC) subsets as a model for autoimmune type 1 diabetes. Our data show that both the BDCA1+ and BDCA3+ mDC subsets engulf mock- as well as virus-infected β-cells, albeit BDCA1+ mDCs are more efficient. Uptake of enterovirus-infected, but not mock-infected cells, activated both DC subsets as indicated by the induction of co-stimulatory molecules and secretion of type I and type III interferons. Both subsets produced similar amounts of interferon-α, yet the BDCA3+ DC were superior in IFN-λ production. The BDCA1+ mDCs more strongly upregulated PD-L1, and were superior in IL-12 and IL-10 production as compared to the BDCA3+ DC. Despite lack of IL-12 production by the BDCA3+ DC, both BDCA1+ and BDCA3+ DCs activated T cells in allogeneic mixed lymphocyte reaction towards a Th1-type reactivity while suppressing Th2-associated cytokines.
Collapse
Affiliation(s)
- Barbara M. Schulte
- Department of Tumor Immunology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Paul R. Gielen
- Department of Tumor Immunology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Esther D. Kers-Rebel
- Department of Tumor Immunology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Frank J. M. van Kuppeveld
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Gosse J. Adema
- Department of Tumor Immunology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
- * E-mail:
| |
Collapse
|
148
|
Larsson PG, Lakshmikanth T, Laitinen OH, Utorova R, Jacobson S, Oikarinen M, Domsgen E, Koivunen MRL, Chaux P, Devard N, Lecouturier V, Almond J, Knip M, Hyöty H, Flodström-Tullberg M. A preclinical study on the efficacy and safety of a new vaccine against Coxsackievirus B1 reveals no risk for accelerated diabetes development in mouse models. Diabetologia 2015; 58:346-54. [PMID: 25370797 DOI: 10.1007/s00125-014-3436-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Accepted: 10/09/2014] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Enterovirus infections have been implicated in the aetiology of autoimmune type 1 diabetes. A vaccine could be used to test the causal relationship between enterovirus infections and diabetes development. However, the development of a vaccine against a virus suspected to induce an autoimmune disease is challenging, since the vaccine itself might trigger autoimmunity. Another challenge is to select the enterovirus serotypes to target with a vaccine. Here we aimed to evaluate the function and autoimmune safety of a novel non-adjuvanted prototype vaccine to Coxsackievirus serotype B1 (CVB1), a member of the enterovirus genus. METHODS A formalin-inactivated CVB1 vaccine was developed and tested for its immunogenicity and safety in BALB/c and NOD mice. Prediabetic NOD mice were vaccinated, infected with CVB1 or mock-treated to compare the effect on diabetes development. RESULTS Vaccinated mice produced high titres of CVB1-neutralising antibodies without signs of vaccine-related side effects. Vaccinated mice challenged with CVB1 had significantly reduced levels of replicating virus in their blood and the pancreas. Prediabetic NOD mice demonstrated an accelerated onset of diabetes upon CVB1 infection whereas no accelerated disease manifestation or increased production of insulin autoantibodies was observed in vaccinated mice. CONCLUSIONS/INTERPRETATION We conclude that the prototype vaccine is safe and confers protection from infection without accelerating diabetes development in mice. These results encourage the development of a multivalent enterovirus vaccine for human use, which could be used to determine whether enterovirus infections trigger beta cell autoimmunity and type 1 diabetes in humans.
Collapse
Affiliation(s)
- Pär G Larsson
- The Center for Infectious Medicine, Department of Medicine HS, Karolinska Institutet, Karolinska University Hospital Huddinge F59, SE-141 86, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Morgan NG, Leete P, Foulis AK, Richardson SJ. Islet inflammation in human type 1 diabetes mellitus. IUBMB Life 2014; 66:723-34. [PMID: 25504835 DOI: 10.1002/iub.1330] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 11/17/2014] [Indexed: 01/12/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is caused by the selective deletion of pancreatic β-cells in response to an assault mounted within the pancreas by infiltrating immune cells. However, this apparently clear and focussed annunciation conceals a stark reality in which the cellular and molecular events leading to β-cell loss remain poorly understood in humans. This reflects the difficulty of studying these processes in living individuals and the fact that, using pathological specimens, islet inflammation has been analysed in fewer than 200 recent-onset cases of T1DM worldwide, over the past century. Nevertheless, insights have been gained and the composition of the islet infiltrate is being disclosed. This is shown to be primarily lymphocytic in nature, with populations of both CD8+ and CD4+ T cells displaying an autoreactivity against specific islet antigenic peptides. The T cells are often accompanied by influent CD20+ B cells, although new data imply that the proportions of these individual cell types vary and that patients fall into at least two distinct categories having either a hyper-immune (CD20Hi) or a pauci-immune (CD20Lo) phenotype. The overall rate of β-cell decline appears to correlate with these two phenotypes such that hyper-immune patients lose β-cells more quickly and tend to develop disease at an earlier age than those with the pauci-immune profile. In this article, we review the evidence which underpins our current understanding of the aetiology of T1DM and highlight both the established features as well as areas of on-going ambiguity and debate.
Collapse
Affiliation(s)
- Noel G Morgan
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | | | | | | |
Collapse
|
150
|
Precechtelova J, Borsanyiova M, Sarmirova S, Bopegamage S. Type I diabetes mellitus: genetic factors and presumptive enteroviral etiology or protection. J Pathog 2014; 2014:738512. [PMID: 25574400 PMCID: PMC4276674 DOI: 10.1155/2014/738512] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/14/2014] [Accepted: 11/09/2014] [Indexed: 02/06/2023] Open
Abstract
We review type 1 diabetes and host genetic components, as well as epigenetics and viruses associated with type 1 diabetes, with added emphasis on the enteroviruses, which are often associated with triggering the disease. Genus Enterovirus is classified into twelve species of which seven (Enterovirus A, Enterovirus B, Enterovirus C, and Enterovirus D and Rhinovirus A, Rhinovirus B, and Rhinovirus C) are human pathogens. These viruses are transmitted mainly by the fecal-oral route; they may also spread via the nasopharyngeal route. Enterovirus infections are highly prevalent, but these infections are usually subclinical or cause a mild flu-like illness. However, infections caused by enteroviruses can sometimes be serious, with manifestations of meningoencephalitis, paralysis, myocarditis, and in neonates a fulminant sepsis-like syndrome. These viruses are often implicated in chronic (inflammatory) diseases as chronic myocarditis, chronic pancreatitis, and type 1 diabetes. In this review we discuss the currently suggested mechanisms involved in the viral induction of type 1 diabetes. We recapitulate current basic knowledge and definitions.
Collapse
Affiliation(s)
- Jana Precechtelova
- Enterovirus Laboratory, Faculty of Medicine, Slovak Medical University, Limbova 12, 83303 Bratislava, Slovakia
| | - Maria Borsanyiova
- Enterovirus Laboratory, Faculty of Medicine, Slovak Medical University, Limbova 12, 83303 Bratislava, Slovakia
| | - Sona Sarmirova
- Enterovirus Laboratory, Faculty of Medicine, Slovak Medical University, Limbova 12, 83303 Bratislava, Slovakia
| | - Shubhada Bopegamage
- Enterovirus Laboratory, Faculty of Medicine, Slovak Medical University, Limbova 12, 83303 Bratislava, Slovakia
| |
Collapse
|