1
|
Mbani CJ, Morvan C, Nekoua MP, Debuysschere C, Alidjinou EK, Moukassa D, Hober D. Enterovirus Antibodies: Friends and Foes. Rev Med Virol 2024; 34:e70004. [PMID: 39505825 DOI: 10.1002/rmv.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/02/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024]
Abstract
Enteroviruses (EV) initiate replication by binding to their cellular receptors, leading to the uncoating and release of the viral genome into the cytosol of the host cell. Neutralising antibodies (NAbs) binding to epitopes on enteroviral capsid proteins can inhibit this infectious process through several mechanisms of neutralisation in vitro. Fc-mediated antibody effector functions such as antibody-dependent cell-mediated cytotoxicity and antibody-dependent cellular phagocytosis have also been described for some EV. However, antibody binding to virions does not always result in viral neutralisation. Non-neutralising antibodies, or sub-neutralising concentrations of antibodies, can enhance infection of viruses, leading to more severe pathologies. This phenomenon, known as antibody-dependent enhancement (ADE) of infection, has been described in vitro and/or in vivo for EV including poliovirus, coxsackievirus B and EV-A71. It has been shown that ADE of EV infection is mediated by FcγRs expressed by monocytes, macrophages, B lymphocytes and granulocytes. Antibodies play a crucial role in the diagnosis and monitoring of infections. They are valuable markers that have been used to establish a link between enteroviral infection and chronic diseases such as type 1 diabetes. Monoclonal and polyclonal antibodies targeting enteroviral proteins have been developed and shown to be effective to prevent or combat EV infections in vitro and in vivo. In addition, vaccines are under development, and clinical trials of vaccines are underway or have been completed, providing hope for the prevention of diseases due to EV. However, the ADE of the infection should be considered in the development of anti-EV antibodies or safe vaccines.
Collapse
Affiliation(s)
- Chaldam Jespère Mbani
- Laboratoire de Virologie URL3610, Univ. Lille et CHU Lille, Lille, France
- Laboratoire de Biologie Cellulaire et Moléculaire, Faculté des Sciences et Techniques, Université Marien Ngouabi, Brazzaville, Congo
| | - Corentin Morvan
- Laboratoire de Virologie URL3610, Univ. Lille et CHU Lille, Lille, France
| | | | - Cyril Debuysschere
- Laboratoire de Virologie URL3610, Univ. Lille et CHU Lille, Lille, France
| | | | - Donatien Moukassa
- Laboratoire de Biologie Cellulaire et Moléculaire, Faculté des Sciences et Techniques, Université Marien Ngouabi, Brazzaville, Congo
| | - Didier Hober
- Laboratoire de Virologie URL3610, Univ. Lille et CHU Lille, Lille, France
| |
Collapse
|
2
|
Ullah S, Zheng Z, Rahman W, Ullah F, Ullah A, Iqbal MN, Iqbal N, Gao T. A computational approach to fighting type 1 diabetes by targeting 2C Coxsackie B virus protein with flavonoids. PLoS One 2023; 18:e0290576. [PMID: 37647325 PMCID: PMC10468086 DOI: 10.1371/journal.pone.0290576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/11/2023] [Indexed: 09/01/2023] Open
Abstract
Autoimmune diabetes, well-known as type 1 insulin-dependent diabetic mellitus (T1D). T1D is a prolonged condition marked by an inadequate supply of insulin. The lack is brought on by pancreatic cell death and results in hyperglycemia. The immune system, genetic predisposition, and environmental variables are just a few of the many elements that contribute significantly to the pathogenicity of T1D disease. In this study, we test flavonoids against Coxsackie virus protein to cope the type 1 diabetes. After protein target identification we perform molecular docking of flavonoids and selected target (1z8r). then performed the ADMET analysis and select the top compound the base of the docking score and the ADMET test analysis. Following that molecular dynamics simulation was performed up to 300 ns. Root means square deviation, root mean square fluctuation, secondary structure elements, and protein-ligand contacts were calculated as post-analysis of simulation. We further check the binding of the ligand with protein by performing MM-GBSA every 10 ns. Lead compound CID_5280445 was chosen as a possible medication based on analysis. The substance is non-toxic, meets the ADMET and BBB likeness requirements, and has the best interaction energy. This work will assist researchers in developing medicine and testing it as a treatment for Diabetes Mellitus Type 1 brought on by Coxsackie B4 viruses by giving them an understanding of chemicals against these viruses.
Collapse
Affiliation(s)
| | - Zilong Zheng
- Big Data Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, P. R. China
| | | | | | - Anees Ullah
- S Khan Lab Mardan, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Nasir Iqbal
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Naveed Iqbal
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Tianshun Gao
- Big Data Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, P. R. China
| |
Collapse
|
3
|
Chen XH, Liu HQ, Nie Q, Wang H, Xiang T. Causal relationship between type 1 diabetes mellitus and six high-frequency infectious diseases: A two-sample mendelian randomization study. Front Endocrinol (Lausanne) 2023; 14:1135726. [PMID: 37065754 PMCID: PMC10102543 DOI: 10.3389/fendo.2023.1135726] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
Purpose Type 1 diabetes mellitus (T1DM) is associated with different types of infections; however, studies on the causal relationship between T1DM and infectious diseases are lacking. Therefore, our study aimed to explore the causalities between T1DM and six high-frequency infections using a Mendelian randomization (MR) approach. Methods Two-sample MR studies were conducted to explore the causalities between T1DM and six high-frequency infections: sepsis, acute lower respiratory infections (ALRIs), intestinal infections (IIs), infections of the genitourinary tract (GUTIs) in pregnancy, infections of the skin and subcutaneous tissues (SSTIs), and urinary tract infections (UTIs). Data on summary statistics for T1DM and infections were obtained from the European Bioinformatics Institute database, the United Kingdom Biobank, FinnGen biobank, and Medical Research Council Integrative Epidemiology Unit. All data obtained for summary statistics were from European countries. The inverse-variance weighted (IVW) method was employed as the main analysis. Considering the multiple comparisons, statistical significance was set at p< 0.008. If univariate MR analyses found a significant causal association, multivariable MR (MVMR) analyses were performed to adjust body mass index (BMI) and glycated hemoglobin (HbA1c). MVMR-IVW was performed as the primary analysis, and the least absolute shrinkage and selection operator (LASSO) regression and MVMR-Robust were performed as complementary analyses. Results MR analysis showed that susceptibility to IIs increased in patients with T1DM by 6.09% using the IVW-fixed method [odds ratio (OR)=1.0609; 95% confidence interval (CI): 1.0281-1.0947, p=0.0002]. Results were still significant after multiple testing. Sensitivity analyses did not show any significant horizontal pleiotropy or heterogeneity. After adjusting for BMI and HbA1c, MVMR-IVW (OR=1.0942; 95% CI: 1.0666-1.1224, p<0.0001) showed significant outcomes that were consistent with those of LASSO regression and MVMR-Robust. However, no significant causal relationship was found between T1DM and sepsis susceptibility, ALRI susceptibility, GUTI susceptibility in pregnancy, SSTI susceptibility, and UTI susceptibility. Conclusions Our MR analysis genetically predicted increased susceptibility to IIs in T1DM. However, no causality between T1DM and sepsis, ALRIs, GUTIs in pregnancy, SSTIs, or UTIs was found. Larger epidemiological and metagenomic studies are required to further investigate the observed associations between the susceptibility of certain infectious diseases with T1DM.
Collapse
Affiliation(s)
- Xiao-Hong Chen
- Emergency Department, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Hong-Qiong Liu
- Department of Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qiong Nie
- Department of Geriatrics, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Han Wang
- Department of Cardiology, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Tao Xiang
- Emergency Department, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Viruses and Endocrine Diseases. Microorganisms 2023; 11:microorganisms11020361. [PMID: 36838326 PMCID: PMC9967810 DOI: 10.3390/microorganisms11020361] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Viral infections have been frequently associated with physiological and pathological changes in the endocrine system for many years. The numerous early and late endocrine complications reported during the current pandemic of coronavirus disease 2019 (COVID-19) reinforce the relevance of improving our understanding of the impact of viral infections on the endocrine system. Several viruses have been shown to infect endocrine cells and induce endocrine system disturbances through the direct damage of these cells or through indirect mechanisms, especially the activation of the host antiviral immune response, which may lead to the development of local or systemic inflammation or organ-specific autoimmunity. In addition, endocrine disorders may also affect susceptibility to viral infections since endocrine hormones have immunoregulatory functions. This review provides a brief overview of the impact of viral infections on the human endocrine system in order to provide new avenues for the control of endocrine diseases.
Collapse
|
5
|
Gharbi J, Almalki MA, Ben M’hadheb M. The introduction of mutations in the wild type coxsackievirus B3 (CVB3) IRES RNA leads to different levels of in vitro reduced replicative and translation efficiencies. PLoS One 2022; 17:e0274162. [PMID: 36190999 PMCID: PMC9529112 DOI: 10.1371/journal.pone.0274162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 08/24/2022] [Indexed: 11/25/2022] Open
Abstract
Coxsackievirus B3 (CVB3) is a principal causative agent of viral myocarditis, meningitis and pancreatitis. There is no vaccine available for clinical use. It has been demonstrated that the primary molecular determinant of virulence phenotype is located in the 5' UTR of the viral genome. Translation initiation of CVB3 RNA is directed by the IRES element situated in the 5'UTR. In the present study, we analyse the effects of single point mutations introduced in different positions in the domain V of the IRES RNA of CVB3 wild type. We characterize in vitro virus replicative capacitiy and translation efficiency and we test in vivo virulence of different CVB3 mutants produced by the introduction of different mutations in the domain V of IRES by site-directed mutagenesis to abolish its structure. Our results demonstrate that all RNA mutants display different levels of decreased replication and translation initiation efficiency in vitro. The translation defect was correlated with significant reduced viral titer of mutant particles in comparison with the wild type. When inoculated in mice, mutant viruses were checked for inflammation and necrosis.In vitro and in vivo Findings strongly suggest that the most attenuated mutant strain could be considered a candidate for live-attenuated CVB3 vaccine.
Collapse
Affiliation(s)
- Jawhar Gharbi
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
- * E-mail:
| | - Mohammed A. Almalki
- Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Manel Ben M’hadheb
- Virology and Antiviral Strategies Research Unit [UR17/ES30 ViroBiotech], Institute of Biotechnology, University of Monastir, Monastir, Tunisia
| |
Collapse
|
6
|
Nekoua MP, Alidjinou EK, Hober D. Persistent coxsackievirus B infection and pathogenesis of type 1 diabetes mellitus. Nat Rev Endocrinol 2022; 18:503-516. [PMID: 35650334 PMCID: PMC9157043 DOI: 10.1038/s41574-022-00688-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/28/2022] [Indexed: 12/15/2022]
Abstract
Enteroviruses are believed to trigger or accelerate islet autoimmunity in genetically susceptible individuals, thereby resulting in loss of functional insulin-producing β-cells and type 1 diabetes mellitus (T1DM). Although enteroviruses are primarily involved in acute and lytic infections in vitro and in vivo, they can also establish a persistent infection. Prospective epidemiological studies have strongly associated the persistence of enteroviruses, especially coxsackievirus B (CVB), with the appearance of islet autoantibodies and an increased risk of T1DM. CVB can persist in pancreatic ductal and β-cells, which leads to structural or functional alterations of these cells, and to a chronic inflammatory response that promotes recruitment and activation of pre-existing autoreactive T cells and β-cell autoimmune destruction. CVB persistence in other sites, such as the intestine, blood cells and thymus, has been described; these sites could serve as a reservoir for infection or reinfection of the pancreas, and this persistence could have a role in the disturbance of tolerance to β-cells. This Review addresses the involvement of persistent enterovirus infection in triggering islet autoimmunity and T1DM, as well as current strategies to control enterovirus infections for preventing or reducing the risk of T1DM onset.
Collapse
Affiliation(s)
| | | | - Didier Hober
- Laboratoire de Virologie ULR3610, Université de Lille, CHU Lille, Lille, France.
| |
Collapse
|
7
|
Li Y, Zhang W, Zhao R, Zhang X. Advances in oral peptide drug nanoparticles for diabetes mellitus treatment. Bioact Mater 2022; 15:392-408. [PMID: 35386357 PMCID: PMC8958389 DOI: 10.1016/j.bioactmat.2022.02.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 02/18/2022] [Accepted: 02/18/2022] [Indexed: 12/11/2022] Open
Abstract
Peptide drugs play an important role in diabetes mellitus treatment. Oral administration of peptide drugs is a promising strategy for diabetes mellitus because of its convenience and high patient compliance compared to parenteral administration routes. However, there are a series of formidable unfavorable conditions present in the gastrointestinal (GI) tract after oral administration, which result in the low oral bioavailability of these peptide drugs. To overcome these challenges, various nanoparticles (NPs) have been developed to improve the oral absorption of peptide drugs due to their unique in vivo properties and high design flexibility. This review discusses the unfavorable conditions present in the GI tract and provides the corresponding strategies to overcome these challenges. The review provides a comprehensive overview on the NPs that have been constructed for oral peptide drug delivery in diabetes mellitus treatment. Finally, we will discuss the rational application and give some suggestions that can be utilized for the development of oral peptide drug NPs. Our aim is to provide a systemic and comprehensive review of oral peptide drug NPs that can overcome the challenges in GI tract for efficient treatment of diabetes mellitus. •Oral administration of peptide drugs is a promising strategy for diabetes mellitus treatment •A series of formidable unfavorable conditions in gastrointestinal tract result in the low oral bioavailability of peptide drugs •Nanoparticles can improve the oral bioavailability of peptide drugs
Collapse
Affiliation(s)
- Yan Li
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Wen Zhang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100083, PR China
| | - Ruichen Zhao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China.,School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Xin Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China
| |
Collapse
|
8
|
Hamdi A, Halouani A, Aouf I, Viaene J, Marzouk B, Kraiem J, Jaïdane H, Heyden YV. Cytotoxicity and Antiviral Activities of Haplophyllum tuberculatum Essential Oils, Pure Compounds, and Their Combinations against Coxsackievirus B3 and B4. PLANTA MEDICA 2021; 87:827-835. [PMID: 34293806 DOI: 10.1055/a-1538-5289] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Haplophyllum tuberculatum is a plant commonly used in folk medicine to treat several diseases including vomiting, nausea, infections, rheumatism, and gastric pains. In the current study, H. tuberculatum essential oils, hydrosols, the pure compounds R-(+)-limonene, S-(-)-limonene, and 1-octanol, as well as their combinations R-(+)-limonene/1-octanol and S-(-)-limonene/1-octanol, were screened for their cytotoxicity on HEp-2 cells after 24, 48, and 72 h, and then tested for their activity against Coxsackievirus B3 and B4 (CV-B3 and CV-B4) at 3 different moments: addition of the plant compounds before, after, or together with virus inoculation. Results showed that the samples were more cytotoxic after 72 h than after 24 h or 48 h cell contact. However, the combinations R-(+)-limonene/1-octanol and S-(-)-limonene/1-octanol showed less effect on HEp-2 cells than pure R-(+)-limonene and S-(-)-limonene after 24 h, 48 h, and 72 h. 1-octanol exhibited the highest concentration causing 50% cytotoxicity (CC50) on HEp-2 cells after 24 h (CC50 = 93 µg/mL) and 48 h (CC50 = 83 µg/mL). The antiviral assays showed that the tested samples exhibited potent inhibition of CV-B. IC50 values ranged from 0.66 µg/mL to 28.4 µg/mL. In addition, CV-B3 was more sensitive than CV-B4. Both CV-B strains are more inhibited when cells were pretreated with the plant compounds. The hydrosols have no effect, neither on HEp-2 cells nor on the virus. 1-octanol, S-(-), and R-(+)-limonene/1-octanol had important selectivity indexes over time. Although essential oils had potent antiviral activity, they can be considered for application in the pretreatment of cells. However, 1-octanol and the combinations are within the safety limits, and thus, they can be used as an active natural antiviral agent for CV-B3 and CV-B4 inhibition.
Collapse
Affiliation(s)
- Assia Hamdi
- Laboratory of Chemical, Pharmaceutical and Pharmacological Development of Drugs, Faculty of Pharmacy, University of Monastir, Tunisia
- Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling (FABI), Center for Pharmaceutical Research (CePhaR), Vrije Universiteit Brussel (VUB), Belgium
| | - Aymen Halouani
- Laboratory of Transmissible Diseases and Biologically Active Substances LR99ES27, Faculty of Pharmacy, University of Monastir, Tunisia
| | - Ines Aouf
- Laboratory of Transmissible Diseases and Biologically Active Substances LR99ES27, Faculty of Pharmacy, University of Monastir, Tunisia
| | - Johan Viaene
- Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling (FABI), Center for Pharmaceutical Research (CePhaR), Vrije Universiteit Brussel (VUB), Belgium
| | - Belsem Marzouk
- Laboratory of Chemical, Pharmaceutical and Pharmacological Development of Drugs, Faculty of Pharmacy, University of Monastir, Tunisia
| | - Jamil Kraiem
- Laboratory of Chemical, Pharmaceutical and Pharmacological Development of Drugs, Faculty of Pharmacy, University of Monastir, Tunisia
| | - Hela Jaïdane
- Laboratory of Transmissible Diseases and Biologically Active Substances LR99ES27, Faculty of Pharmacy, University of Monastir, Tunisia
| | - Yvan Vander Heyden
- Department of Analytical Chemistry, Applied Chemometrics and Molecular Modelling (FABI), Center for Pharmaceutical Research (CePhaR), Vrije Universiteit Brussel (VUB), Belgium
| |
Collapse
|
9
|
Halouani A, Michaux H, Jmii H, Trussart C, Chahbi A, Martens H, Renard C, Aouni M, Hober D, Geenen V, Jaïdane H. Coxsackievirus B4 Transplacental Infection Severely Disturbs Central Tolerogenic Mechanisms in the Fetal Thymus. Microorganisms 2021; 9:microorganisms9071537. [PMID: 34361972 PMCID: PMC8303261 DOI: 10.3390/microorganisms9071537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/10/2021] [Accepted: 07/16/2021] [Indexed: 01/09/2023] Open
Abstract
Thymus plays a fundamental role in central tolerance establishment, especially during fetal life, through the generation of self-tolerant T cells. This process consists in T cells education by presenting them tissue-restricted autoantigens promiscuously expressed by thymic epithelial cells (TECs), thus preventing autoimmunity. Thymus infection by Coxsackievirus B (CV-B) during fetal life is supposed to disturb thymic functions and, hence, to be an inducing or accelerating factor in the genesis of autoimmunity. To further investigate this hypothesis, in our current study, we analyzed thymic expression of autoantigens, at the transcriptional and protein level, following in utero infection by CV-B4. mRNA expression levels of Igf2 and Myo7, major autoantigens of pancreas and heart, respectively, were analyzed in whole thymus and in enriched TECs together along with both transcription factors, Aire and Fezf2, involved in autoantigens expression in the thymus. Results show that in utero infection by CV-B4 induces a significant decrease in Igf2 and Myo7 expression at both mRNA and protein level in whole thymus and in enriched TECs as well. Moreover, a correlation between viral load and autoantigens expression can be observed in the whole thymus, indicating a direct effect of in utero infection by CV-B4 on autoantigens expression. Together, these results indicate that an in utero infection of the thymus by CV-B4 may interfere with self-tolerance establishment in TECs by decreasing autoantigen expression at both mRNA and protein level and thereby increase the risk of autoimmunity onset.
Collapse
Affiliation(s)
- Aymen Halouani
- Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir 5000, Tunisia; (A.H.); (H.J.); (M.A.)
- Faculté des Sciences de Tunis, Université de Tunis El Manar, Tunis 1068, Tunisia
- GIGA-I3 Immunoendocrinologie, Faculté de Médicine, Université de Liège, CHU-B34, Sart Tilman, 4000 Liège, Belgium; (H.M.); (C.T.); (H.M.); (C.R.); (V.G.)
| | - Hélène Michaux
- GIGA-I3 Immunoendocrinologie, Faculté de Médicine, Université de Liège, CHU-B34, Sart Tilman, 4000 Liège, Belgium; (H.M.); (C.T.); (H.M.); (C.R.); (V.G.)
| | - Habib Jmii
- Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir 5000, Tunisia; (A.H.); (H.J.); (M.A.)
- Faculté des Sciences de Tunis, Université de Tunis El Manar, Tunis 1068, Tunisia
| | - Charlotte Trussart
- GIGA-I3 Immunoendocrinologie, Faculté de Médicine, Université de Liège, CHU-B34, Sart Tilman, 4000 Liège, Belgium; (H.M.); (C.T.); (H.M.); (C.R.); (V.G.)
| | - Ahlem Chahbi
- Laboratoire d’Hématologie, Faculté de Médecine de Tunis, Université de Tunis El Manar, Tunis 1007, Tunisia;
| | - Henri Martens
- GIGA-I3 Immunoendocrinologie, Faculté de Médicine, Université de Liège, CHU-B34, Sart Tilman, 4000 Liège, Belgium; (H.M.); (C.T.); (H.M.); (C.R.); (V.G.)
| | - Chantal Renard
- GIGA-I3 Immunoendocrinologie, Faculté de Médicine, Université de Liège, CHU-B34, Sart Tilman, 4000 Liège, Belgium; (H.M.); (C.T.); (H.M.); (C.R.); (V.G.)
| | - Mahjoub Aouni
- Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir 5000, Tunisia; (A.H.); (H.J.); (M.A.)
| | - Didier Hober
- Laboratoire de Virologie EA3610, Faculté de Médecine, Université de Lille, CHU Lille, 59000 Lille, France;
| | - Vincent Geenen
- GIGA-I3 Immunoendocrinologie, Faculté de Médicine, Université de Liège, CHU-B34, Sart Tilman, 4000 Liège, Belgium; (H.M.); (C.T.); (H.M.); (C.R.); (V.G.)
| | - Hela Jaïdane
- Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir, Université de Monastir, Monastir 5000, Tunisia; (A.H.); (H.J.); (M.A.)
- Correspondence: ; Tel.: +216-98-677-174
| |
Collapse
|
10
|
Innate immune receptors in type 1 diabetes: the relationship to cell death-associated inflammation. Biochem Soc Trans 2021; 48:1213-1225. [PMID: 32510139 DOI: 10.1042/bst20200131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/17/2022]
Abstract
The importance of innate immunity in host defense and inflammatory responses has been clearly demonstrated after the discovery of innate immune receptors such as Toll-like receptors (TLRs) or Nucleotide-binding oligomerization domain-containing protein (Nod)-like receptors (NLRs). Innate immunity also plays a critical role in diverse pathological conditions including autoimmune diseases such as type 1 diabetes (T1D). In particular, the role of a variety of innate immune receptors in T1D has been demonstrated using mice with targeted disruption of such innate immune receptors. Here, we discuss recent findings showing the role of innate immunity in T1D that were obtained mostly from studies of genetic mouse models of innate immune receptors. In addition, the role of innate immune receptors involved in the pathogenesis of T1D in sensing death-associated molecular patterns (DAMPs) released from dead cells or pathogen-associated molecular patterns (PAMPs) will also be covered. Elucidation of the role of innate immune receptors in T1D and the nature of DAMPs sensed by such receptors may lead to the development of new therapeutic modalities against T1D.
Collapse
|
11
|
Real-Fernández F, Gallo A, Nuti F, Altamore L, Del Vescovo GG, Traldi P, Ragazzi E, Rovero P, Lapolla A, Papini AM. Cross-reactive peptide epitopes of Enterovirus Coxsackie B4 and human glutamic acid decarboxylase detecting antibodies in latent autoimmune diabetes in adults versus type 1 diabetes. Clin Chim Acta 2021; 515:73-79. [PMID: 33422493 DOI: 10.1016/j.cca.2021.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/31/2020] [Accepted: 01/05/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Diagnosis of latent autoimmune diabetes in adults (LADA) is usually based on the adult age, anti-pancreatic islet cell antibodies detection, and insulin independence. This study investigates the diagnostic value of antibodies against human glutamic acid decarboxylase (hGAD) peptides in LADA and type 1 diabetes mellitus (T1DM) patients, and their cross-reactivity with an Enterovirus Coxsackie B4 (CVB4) shared epitope. METHODS Sera from 27 LADA patients, 23 T1DM patients, and 24 controls were tested in ELISA for antibodies against hGAD peptides and a selected sequence of P2C protein of CVB4 (CVB4P2C). Diagnostic power of peptides was analyzed by ROC-curve analysis and cross-reactivity among peptides evaluated. RESULTS IgM and IgG antibodies showed significant differences between LADA and T1DM versus controls for all peptides. Antibody responses present high agreement among peptides for IgM and IgG-isotypes in T1DM, which is not reproduced in LADA. IgM antibodies showed high predicting diagnostic power particularly in LADA (sensitivity > 85%, specificity 95.8%). CONCLUSIONS Our study highlights the usefulness of peptides as diagnostic antigens in T1DM and LADA, and extends previous findings by comparing IgM and IgG-isotype antibodies in the same population. Additionally, results highlight the role of the entourage in the shared sequon PEVKXK in GAD and CVB4P2C particularly in IgMs identification.
Collapse
Affiliation(s)
- Feliciana Real-Fernández
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Alessandra Gallo
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, Padova, Italy
| | - Francesca Nuti
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Lorenzo Altamore
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | | | - Pietro Traldi
- Istituto di Ricerca Pediatrica, Città della Speranza, Padova, Italy
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, School of Medicine and Surgery, University of Padova, Padova, Italy
| | - Paolo Rovero
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of Neurosciences, Psychology, Drug Research and Child Health, Section of Pharmaceutical Sciences and Nutraceutics, University of Florence, Sesto Fiorentino, Italy
| | - Annunziata Lapolla
- Diabetology and Dietetics, Department of Medicine (DIMED), University of Padova, Padova, Italy.
| | - Anna Maria Papini
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy; PeptLab@UCP, CY Cergy Paris Université, Cergy Pontoise, France.
| |
Collapse
|
12
|
Elhag DA, Kumar M, Al Khodor S. Exploring the Triple Interaction between the Host Genome, the Epigenome, and the Gut Microbiome in Type 1 Diabetes. Int J Mol Sci 2020; 22:ijms22010125. [PMID: 33374418 PMCID: PMC7795494 DOI: 10.3390/ijms22010125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/13/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Type 1 diabetes (T1D) is an auto-immune disorder characterized by a complex interaction between the host immune system and various environmental factors in genetically susceptible individuals. Genome-wide association studies (GWAS) identified different T1D risk and protection alleles, however, little is known about the environmental factors that can be linked to these alleles. Recent evidence indicated that, among those environmental factors, dysbiosis (imbalance) in the gut microbiota may play a role in the pathogenesis of T1D, affecting the integrity of the gut and leading to systemic inflammation and auto-destruction of the pancreatic β cells. Several studies have identified changes in the gut microbiome composition in humans and animal models comparing T1D subjects with controls. Those changes were characterized by a higher abundance of Bacteroides and a lower abundance of the butyrate-producing bacteria such as Clostridium clusters IV and XIVa. The mechanisms by which the dysbiotic bacteria and/or their metabolites interact with the genome and/or the epigenome of the host leading to destructive autoimmunity is still not clear. As T1D is a multifactorial disease, understanding the interaction between different environmental factors such as the gut microbiome, the genetic and the epigenetic determinants that are linked with the early appearance of autoantibodies can expand our knowledge about the disease pathogenesis. This review aims to provide insights into the interaction between the gut microbiome, susceptibility genes, epigenetic factors, and the immune system in the pathogenesis of T1D.
Collapse
|
13
|
Honkimaa A, Kimura B, Sioofy-Khojine AB, Lin J, Laiho J, Oikarinen S, Hyöty H. Genetic Adaptation of Coxsackievirus B1 during Persistent Infection in Pancreatic Cells. Microorganisms 2020; 8:microorganisms8111790. [PMID: 33203081 PMCID: PMC7697981 DOI: 10.3390/microorganisms8111790] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/16/2022] Open
Abstract
Coxsackie B (CVB) viruses have been associated with type 1 diabetes. We have recently observed that CVB1 was linked to the initiation of the autoimmune process leading to type 1 diabetes in Finnish children. Viral persistency in the pancreas is currently considered as one possible mechanism. In the current study persistent infection was established in pancreatic ductal and beta cell lines (PANC-1 and 1.1B4) using four different CVB1 strains, including the prototype strain and three clinical isolates. We sequenced 5′ untranslated region (UTR) and regions coding for structural and non-structural proteins and the second single open reading frame (ORF) protein of all persisting CVB1 strains using next generation sequencing to identify mutations that are common for all of these strains. One mutation, K257R in VP1, was found from all persisting CVB1 strains. The mutations were mainly accumulated in viral structural proteins, especially at BC, DE, EF loops and C-terminus of viral capsid protein 1 (VP1), the puff region of VP2, the knob region of VP3 and infection-enhancing epitope of VP4. This showed that the capsid region of the viruses sustains various changes during persistency some of which could be hallmark(s) of persistency.
Collapse
Affiliation(s)
- Anni Honkimaa
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (B.K.); (A.B.S.-K.); (J.L.); (S.O.); (H.H.)
- Correspondence:
| | - Bryn Kimura
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (B.K.); (A.B.S.-K.); (J.L.); (S.O.); (H.H.)
| | - Amir-Babak Sioofy-Khojine
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (B.K.); (A.B.S.-K.); (J.L.); (S.O.); (H.H.)
| | - Jake Lin
- Finnish Institute of Molecular Medicine (FIMM), University of Helsinki, 00290 Helsinki, Finland;
| | - Jutta Laiho
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (B.K.); (A.B.S.-K.); (J.L.); (S.O.); (H.H.)
| | - Sami Oikarinen
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (B.K.); (A.B.S.-K.); (J.L.); (S.O.); (H.H.)
| | - Heikki Hyöty
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (B.K.); (A.B.S.-K.); (J.L.); (S.O.); (H.H.)
- Fimlab Laboratories, Pirkanmaa Hospital District, 33520 Tampere, Finland
| |
Collapse
|
14
|
Enteroviral Pathogenesis of Type 1 Diabetes: The Role of Natural Killer Cells. Microorganisms 2020; 8:microorganisms8070989. [PMID: 32630332 PMCID: PMC7409131 DOI: 10.3390/microorganisms8070989] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 12/16/2022] Open
Abstract
Enteroviruses, especially group B coxsackieviruses (CV-B), have been associated with the development of chronic diseases such as type 1 diabetes (T1D). The pathological mechanisms that trigger virus-induced autoimmunity against islet antigens in T1D are not fully elucidated. Animal and human studies suggest that NK cells response to CV-B infection play a crucial role in the enteroviral pathogenesis of T1D. Indeed, CV-B-infected cells can escape from cytotoxic T cells recognition and destruction by inhibition of cell surface expression of HLA class I antigen through non-structural viral proteins, but they can nevertheless be killed by NK cells. Cytolytic activity of NK cells towards pancreatic beta cells persistently-infected with CV-B has been reported and defective viral clearance by NK cells of patients with T1D has been suggested as a mechanism leading to persistence of CV-B and triggering autoimmunity reported in these patients. The knowledge about host antiviral defense against CV-B infection is not only crucial to understand the susceptibility to virus-induced T1D but could also contribute to the design of new preventive or therapeutic approaches for individuals at risk for T1D or newly diagnosed patients.
Collapse
|
15
|
Honkimaa A, Sioofy-Khojine AB, Oikarinen S, Bertin A, Hober D, Hyöty H. Eradication of persistent coxsackievirus B infection from a pancreatic cell line with clinically used antiviral drugs. J Clin Virol 2020; 128:104334. [PMID: 32450550 DOI: 10.1016/j.jcv.2020.104334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 03/06/2020] [Accepted: 03/23/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Persistent enterovirus infections create a difficult therapeutic challenge in immunocompromised patients and may also contribute to the development of chronic diseases including type 1 diabetes, cardiomyopathies, post-polio syndrome and chronic fatigue syndrome. OBJECTIVES To study the ability of antiviral drugs to eradicate such infection in vitro to evalaute their potential in the treatments of these patients. STUDY DESIGN We set out to evaluate several licensed or clinically tested drugs which have shown some anti-enterovirus activity in previous studies for their ability to cure persistent infection established by two different coxsackievirus B1 strains in a pancreatic cell line (PANC-1 cells). RESULTS Among all tested drugs Enviroxime, Fluoxetine, concentrated human IgG product (Hizentra) and Pleconaril were able to eradicate persistent Coxsackievirus B1 infection. The effect Enviroxime, Hizentra and Pleconaril varied between the two virus strains. CONCLUSIONS The identified drugs are feasible candidates for clinical trials among patients with persistent coxsackievirus B infections or chronic enterovirus-associated diseases.
Collapse
Affiliation(s)
- Anni Honkimaa
- Tampere University, Faculty of Medicine and Health Technology, Arvo Ylpönkatu 34, FIN-33520 Tampere, Finland.
| | - Amir-Babak Sioofy-Khojine
- Tampere University, Faculty of Medicine and Health Technology, Arvo Ylpönkatu 34, FIN-33520 Tampere, Finland
| | - Sami Oikarinen
- Tampere University, Faculty of Medicine and Health Technology, Arvo Ylpönkatu 34, FIN-33520 Tampere, Finland
| | - Antoine Bertin
- Université de Lille, CHU Lille Laboratoire de Virologie, EA3610, F-59000 Lille, France
| | - Didier Hober
- Université de Lille, CHU Lille Laboratoire de Virologie, EA3610, F-59000 Lille, France
| | - Heikki Hyöty
- Tampere University, Faculty of Medicine and Health Technology, Arvo Ylpönkatu 34, FIN-33520 Tampere, Finland; Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| |
Collapse
|
16
|
Immunopathology in the brain of mice following vertical transmission of Coxsackievirus B4. Microb Pathog 2020; 140:103965. [PMID: 31904449 DOI: 10.1016/j.micpath.2020.103965] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 01/01/2020] [Indexed: 12/15/2022]
Abstract
Coxsackie B viruses (CV-B) are associated with several central nervous system (CNS) disorders. These viruses are predominantly transmitted by fecal-oral route but vertical transmission can also occur. This work attempted to study the immune response ensuing vertical transmission of CV-B to the brain, and its eventual implementation in the brain pathogenesis. To this end, pregnant Swiss albino mice were inoculated with CV-B4 E2 or with sterile medium for control animals. At different ages after birth, brains were collected and analyzed for virus infection, histopathological changes and immune response. Infectious particles were detected in offspring's brain which demonstrates vertical transmission of the virus. This infection is persistent since the long lasting detection of viral RNA in offspring's brain. Some pathological signs including meningitis, edema and accumulation of inflammatory cells within and surrounding the inflammatory areas were observed. Immunoflorescence staining unveiled the presence of T lymphocytes and microgliosis in the sites of lesion for a long period after birth. Multiplex cytokines measurement upon supernatants of in vitro mixed brain cells and extracted mononuclear cells from offspring's brain has demonstrated an elevated secretion of the pro-inflammatory cytokines TNFα, IL-6 and IFNα and the chemokines RANTES and MCP-1. Hence, vertical transmission of CV-B4 and its persistence within offspring's brain can lead to pathological features linked to increased and sustained immune response.
Collapse
|
17
|
Coxsackievirus-B4 Infection of Human Primary Pancreatic Ductal Cell Cultures Results in Impairment of Differentiation into Insulin-Producing Cells. Viruses 2019; 11:v11070597. [PMID: 31269669 PMCID: PMC6669621 DOI: 10.3390/v11070597] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/11/2019] [Accepted: 06/27/2019] [Indexed: 12/15/2022] Open
Abstract
Coxsackievirus-B4 (CV-B4) E2 can persist in the pancreatic ductal-like cells (Panc-1 cell line), which results in an impaired differentiation of these cells into islet-like cell aggregates (ICA). In this study, primary pancreatic ductal cells obtained as a by-product of islet isolation from the pancreas of seven brain-dead adults were inoculated with CV-B4 E2, followed-up for 29 days, and the impact was investigated. Viral titers in culture supernatants were analyzed throughout the culture. Intracellular viral RNA was detected by RT-PCR. Levels of ductal cell marker CK19 mRNA and of insulin mRNA were evaluated by qRT-PCR. The concentration of c-peptide in supernatants was determined by ELISA. Ductal cells exposed to trypsin and serum-free medium formed ICA and resulted in an increased insulin secretion. Ductal cells from five brain-dead donors were severely damaged by CV-B4 E2, whereas the virus persisted in cultures of cells obtained from the other two. The ICAs whose formation was induced on day 14 post-inoculation were scarce and appeared tiny in infected cultures. Also, insulin mRNA expression and c-peptide levels were strongly reduced compared to the controls. In conclusion, CV-B4 E2 lysed human primary pancreatic ductal cells or persisted in these cells, which resulted in the impairment of differentiation into insulin-producing cells.
Collapse
|
18
|
Nekoua MP, Yessoufou A, Alidjinou EK, Badia-Boungou F, Moutairou K, Sane F, Hober D. Salivary anti-coxsackievirus-B4 neutralizing activity and pattern of immune parameters in patients with type 1 diabetes: a pilot study. Acta Diabetol 2018; 55:827-834. [PMID: 29774468 DOI: 10.1007/s00592-018-1158-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/06/2018] [Indexed: 12/15/2022]
Abstract
AIMS Enteroviruses, especially coxsackieviruses B (CV-B), have been associated with the pathogenesis of type 1 diabetes (T1D). An anti-CV-B4 neutralizing activity in saliva of T1D patients was previously reported. Our aim was to study the association between the saliva anti-CV-B4 neutralizing activity and immune parameters in T1D patients in comparison with non-diabetic individuals. METHODS Saliva and blood samples were collected from 15 T1D patients and 8 controls. The anti-CV-B4 and anti-poliovirus type 1 (PV-1) activities of saliva and serum samples were determined by a plaque neutralization assay. Quantification of serum cytokines was performed by ELISA and the frequencies of lymphocyte subsets were evaluated using flow cytometry. RESULTS The levels of salivary anti-CV-B4 neutralizing activity were higher in T1D patients than in controls (p = 0.02), whereas the serum levels of anti-CV-B4 neutralizing activity and the saliva and serum levels of anti-PV-1 neutralizing activity were not different. The proportions of effector CD4+ T cells and CD19+ B cells, but not those of CD4+ T cells, CD8+ T cells and Foxp3+ regulatory T cells, were higher in T1D patients than in controls (p = 0.02 and p = 0.01 respectively). Moreover, serum IFN-γ levels were lower in T1D patients compared to controls (p = 0.03) while IL-4 and IL-10 were not different. There was an association between saliva anti-CV-B4 activity, down-regulation of IFN-γ and B cell expansion in peripheral blood of T1D patients. CONCLUSION The association between saliva anti-CV-B4 activity and disturbance of immune system in T1D patients deserves further investigation.
Collapse
Affiliation(s)
- Magloire Pandoua Nekoua
- Université de Lille, Faculté de Médecine, CHU de Lille, Laboratoire de Virologie EA3610, 59000, Lille, France
- Université d'Abomey-Calavi, Faculté des Sciences et Techniques, Institut des Sciences Biomédicales Appliquées (ISBA), Laboratoire de Biologie et Physiologie Cellulaires, 01 BP 526, Cotonou, Benin
| | - Akadiri Yessoufou
- Université d'Abomey-Calavi, Faculté des Sciences et Techniques, Institut des Sciences Biomédicales Appliquées (ISBA), Laboratoire de Biologie et Physiologie Cellulaires, 01 BP 526, Cotonou, Benin
| | - Enagnon Kazali Alidjinou
- Université de Lille, Faculté de Médecine, CHU de Lille, Laboratoire de Virologie EA3610, 59000, Lille, France
| | - Francis Badia-Boungou
- Université de Lille, Faculté de Médecine, CHU de Lille, Laboratoire de Virologie EA3610, 59000, Lille, France
| | - Kabirou Moutairou
- Université d'Abomey-Calavi, Faculté des Sciences et Techniques, Institut des Sciences Biomédicales Appliquées (ISBA), Laboratoire de Biologie et Physiologie Cellulaires, 01 BP 526, Cotonou, Benin
| | - Famara Sane
- Université de Lille, Faculté de Médecine, CHU de Lille, Laboratoire de Virologie EA3610, 59000, Lille, France
| | - Didier Hober
- Université de Lille, Faculté de Médecine, CHU de Lille, Laboratoire de Virologie EA3610, 59000, Lille, France.
| |
Collapse
|
19
|
Laitinen OH, Svedin E, Kapell S, Hankaniemi MM, Larsson PG, Domsgen E, Stone VM, Määttä JAE, Hyöty H, Hytönen VP, Flodström-Tullberg M. New Coxsackievirus 2A pro and 3C pro protease antibodies for virus detection and discovery of pathogenic mechanisms. J Virol Methods 2018; 255:29-37. [PMID: 29425680 DOI: 10.1016/j.jviromet.2018.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/01/2018] [Accepted: 02/01/2018] [Indexed: 12/16/2022]
Abstract
Enteroviruses (EVs), such as the Coxsackie B-viruses (CVBs), are common human pathogens, which can cause severe diseases including meningitis, myocarditis and neonatal sepsis. EVs encode two proteases (2Apro and 3Cpro), which perform the proteolytic cleavage of the CVB polyprotein and also cleave host cell proteins to facilitate viral replication. The 2Apro cause direct damage to the infected heart and tools to investigate 2Apro and 3Cpro expression may contribute new knowledge on virus-induced pathologies. Here, we developed new antibodies to CVB-encoded 2Apro and 3Cpro; Two monoclonal 2Apro antibodies and one 3Cpro antibody were produced. Using cells infected with selected viruses belonging to the EV A, B and C species and immunocytochemistry, we demonstrate that the 3Cpro antibody detects all of the EV species B (EV-B) viruses tested and that the 2Apro antibody detects all EV-B viruses apart from Echovirus 9. We furthermore show that the new antibodies work in Western blotting, immunocyto- and immunohistochemistry, and flow cytometry to detect CVBs. Confocal microscopy demonstrated the expression kinetics of 2Apro and 3Cpro, and revealed a preferential cytosolic localization of the proteases in CVB3 infected cells. In summary, the new antibodies detect proteases that belong to EV species B in cells and tissue using multiple applications.
Collapse
Affiliation(s)
- Olli H Laitinen
- The Center for Infectious Medicine, Department of Medicine HS, Karolinska Institutet, Karolinska University Hospital, Stockholm, 141 86, Sweden
| | - Emma Svedin
- The Center for Infectious Medicine, Department of Medicine HS, Karolinska Institutet, Karolinska University Hospital, Stockholm, 141 86, Sweden
| | - Sebastian Kapell
- The Center for Infectious Medicine, Department of Medicine HS, Karolinska Institutet, Karolinska University Hospital, Stockholm, 141 86, Sweden
| | - Minna M Hankaniemi
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33520, Finland; Fimlab Laboratories, 33520 Tampere, Finland
| | - Pär G Larsson
- The Center for Infectious Medicine, Department of Medicine HS, Karolinska Institutet, Karolinska University Hospital, Stockholm, 141 86, Sweden
| | - Erna Domsgen
- The Center for Infectious Medicine, Department of Medicine HS, Karolinska Institutet, Karolinska University Hospital, Stockholm, 141 86, Sweden
| | - Virginia M Stone
- The Center for Infectious Medicine, Department of Medicine HS, Karolinska Institutet, Karolinska University Hospital, Stockholm, 141 86, Sweden
| | - Juha A E Määttä
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33520, Finland; Fimlab Laboratories, 33520 Tampere, Finland
| | - Heikki Hyöty
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33520, Finland; Fimlab Laboratories, 33520 Tampere, Finland
| | - Vesa P Hytönen
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33520, Finland; Fimlab Laboratories, 33520 Tampere, Finland
| | - Malin Flodström-Tullberg
- The Center for Infectious Medicine, Department of Medicine HS, Karolinska Institutet, Karolinska University Hospital, Stockholm, 141 86, Sweden; Faculty of Medicine and Life Sciences, University of Tampere, Tampere, 33520, Finland.
| |
Collapse
|
20
|
Arena MP, Elmastour F, Sane F, Drider D, Fiocco D, Spano G, Hober D. Inhibition of coxsackievirus B4 by Lactobacillus plantarum. Microbiol Res 2018; 210:59-64. [DOI: 10.1016/j.micres.2018.03.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/05/2018] [Accepted: 03/17/2018] [Indexed: 01/14/2023]
|
21
|
Aguech-Oueslati L, Jaidane H, Sane F, Jrad-Battikh N, Hamed SB, Hober D, Gharbi J. Evaluation of Contamination Risks with Coxsackievirus B4 E2 in Swiss Albino Mice Stools. Curr Microbiol 2018; 75:32-39. [PMID: 28856411 DOI: 10.1007/s00284-017-1347-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 08/23/2017] [Indexed: 12/16/2022]
Abstract
Coxsackie B4 (CV-B4), is a major cause of viral myocarditis, dilated cardiomyopathy, and pancreatitis. Like other human enteroviruses, CV-B4 is ubiquitous, excreted in the stool, transmitted by fecal-oral route, and persists in the environment. In the context of studies on CV-B4 infection, it is important to investigate how this virus can be eliminated and to show the possibility of contamination risk with a CV-B4 E2 infected Swiss albino mice. Swiss albino female mice were inoculated with CV-B4 E2 strain and divided in two groups: the first was intraperitoneally (I.P.) infected; the second was orally infected. In order to study the CV-B4 E2 infection in mice, total RNA was extracted from thymus, spleen, pancreas, and intestine, and viral genome was detected using semi-nested (RT-PCR). To further demonstrate infection or immunization of mice, Sera obtained from infected mice were assayed in vitro for their neutralizing antibody. To detect virus in stool of infected mice, stool samples were collected at different post-infection (p.i.) times. Neutralizing antibodies were detectable all along the follow-up period (Day 0, 1, 3, 7, 9, 17, 22, 30, 45, 60 p.i.) in I.P and oral infected mice. Our results showed that when mice were inoculated successively at day 0 and day 8, neutralizing activity was increased in I.P route more than in the oral route. Viral isolation in HEp-2 cells showed negative results. Stool viral analyses reveal a low detection of the CV-B4 E2 genome for all infected mice. In conclusion, our experiments demonstrated that there are no risks linked with the stool of CV-B4 E2 of Swiss albino mice. It would be interesting to characterize the inhibitors of the virus infectivity in these biological samples (stool) and investigate their targets and mechanisms of action.
Collapse
Affiliation(s)
- Leïla Aguech-Oueslati
- Unité de Recherche UR17ES30 "Génomique, Biotechnologie et Stratégies Antivirales»", Institut Supérieur de Biotechnologie, Université de Monastir, BP74, Avenue Tahar Hadded, 5000, Monastir, Tunisia
- et CHU de Lille Laboratoire de Virologie, Université de Lille, EA3610, 59037, Lille, France
| | - Hela Jaidane
- Unité de Recherche UR17ES30 "Génomique, Biotechnologie et Stratégies Antivirales»", Institut Supérieur de Biotechnologie, Université de Monastir, BP74, Avenue Tahar Hadded, 5000, Monastir, Tunisia
- et CHU de Lille Laboratoire de Virologie, Université de Lille, EA3610, 59037, Lille, France
| | - Famara Sane
- et CHU de Lille Laboratoire de Virologie, Université de Lille, EA3610, 59037, Lille, France
| | - Nedia Jrad-Battikh
- Unité de Recherche UR17ES30 "Génomique, Biotechnologie et Stratégies Antivirales»", Institut Supérieur de Biotechnologie, Université de Monastir, BP74, Avenue Tahar Hadded, 5000, Monastir, Tunisia
| | - Sabrine Ben Hamed
- Unité de Recherche UR17ES30 "Génomique, Biotechnologie et Stratégies Antivirales»", Institut Supérieur de Biotechnologie, Université de Monastir, BP74, Avenue Tahar Hadded, 5000, Monastir, Tunisia
| | - Didier Hober
- et CHU de Lille Laboratoire de Virologie, Université de Lille, EA3610, 59037, Lille, France
| | - Jawhar Gharbi
- Unité de Recherche UR17ES30 "Génomique, Biotechnologie et Stratégies Antivirales»", Institut Supérieur de Biotechnologie, Université de Monastir, BP74, Avenue Tahar Hadded, 5000, Monastir, Tunisia.
| |
Collapse
|
22
|
Paschou SA, Papadopoulou-Marketou N, Chrousos GP, Kanaka-Gantenbein C. On type 1 diabetes mellitus pathogenesis. Endocr Connect 2018; 7:R38-R46. [PMID: 29191919 PMCID: PMC5776665 DOI: 10.1530/ec-17-0347] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 11/30/2017] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes mellitus (T1DM) results from the autoimmune destruction of β cells of the endocrine pancreas. Pathogenesis of T1DM is different from that of type 2 diabetes mellitus, where both insulin resistance and reduced secretion of insulin by the β cells play a synergistic role. We will present genetic, environmental and immunologic factors that destroy β cells of the endocrine pancreas and lead to insulin deficiency. The process of autoimmune destruction takes place in genetically susceptible individuals under the triggering effect of one or more environmental factors and usually progresses over a period of many months to years, during which period patients are asymptomatic and euglycemic, but positive for relevant autoantibodies. Symptomatic hyperglycemia and frank diabetes occur after a long latency period, which reflects the large percentage of β cells that need to be destroyed before overt diabetes become evident.
Collapse
Affiliation(s)
- Stavroula A Paschou
- Division of EndocrinologyMetabolism and Diabetes, First Department of Pediatrics, 'Aghia Sophia' Children's Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nektaria Papadopoulou-Marketou
- Division of EndocrinologyMetabolism and Diabetes, First Department of Pediatrics, 'Aghia Sophia' Children's Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - George P Chrousos
- Division of EndocrinologyMetabolism and Diabetes, First Department of Pediatrics, 'Aghia Sophia' Children's Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Kanaka-Gantenbein
- Division of EndocrinologyMetabolism and Diabetes, First Department of Pediatrics, 'Aghia Sophia' Children's Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
23
|
Shen H, Liu T, Luo Y, Shao S, Deng X, Wang H. Echovirus plays major roles in the natural recombination of Coxsackievirus B3. J Med Virol 2017; 90:377-382. [PMID: 28851122 DOI: 10.1002/jmv.24929] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/27/2017] [Indexed: 11/11/2022]
Abstract
Coxsakievirus B3 (CVB3) is a member of enterovirus B (EVB) group, which can cause serious heart diseases such as viral myocarditis. In order to analyze the evolution of CVB3, we performed a recombination analysis of all viral genomes of enterovirus B, and found that there were 19 putative recombination events that produced CVB3. A total of 11 serotypes were found to be involved in the generation of CVB3 progeny virus. These recombination events involved echovirus, EcoV (which includes EcoV6, EcoV9, EcoV14, EcoV15, EcoV17, EcoV21, EcoV24, and EcoV25), CVB4, CVB5, and EVB81, as major or minor parents. The most active, EcoV, which was involved in the 14 of 19 recombination events, acts as one of the parental viruses for CVB3 strains among molecular evolution and recombination events in circulating CVB3. Our study indicates that, EcoV plays major roles in CVB3 recombination, and is involved in the production of 11 new CVB3 recombinant strains.
Collapse
Affiliation(s)
- Hongxing Shen
- Medical College, Jiangsu University, Zhenjiang, P.R. China
| | - Tingjun Liu
- Medical College, Jiangsu University, Zhenjiang, P.R. China
| | - Yucheng Luo
- People's Hospital of Xinghua, Xinghua, P.R. China
| | - Shihe Shao
- Medical College, Jiangsu University, Zhenjiang, P.R. China
| | - Xintao Deng
- People's Hospital of Xinghua, Xinghua, P.R. China
| | - Hua Wang
- Medical College, Jiangsu University, Zhenjiang, P.R. China
| |
Collapse
|
24
|
Engelmann I, Alidjinou EK, Bertin A, Bossu J, Villenet C, Figeac M, Sane F, Hober D. Persistent coxsackievirus B4 infection induces microRNA dysregulation in human pancreatic cells. Cell Mol Life Sci 2017; 74:3851-3861. [PMID: 28601984 PMCID: PMC11107484 DOI: 10.1007/s00018-017-2567-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/16/2017] [Accepted: 06/06/2017] [Indexed: 12/15/2022]
Abstract
Enterovirus infections are implicated in the development of type 1 diabetes (T1D). MicroRNAs as regulators of gene expression are involved in many physiological and pathological processes. Given that viral infections dysregulate cellular microRNAs, we investigated the impact of persistent coxsackievirus B4 infection on microRNA expression of human pancreatic cells. Next-generation sequencing was used to determine microRNA expression in PANC-1 cells persistently infected (for several weeks) with coxsackievirus B4 and uninfected control cells. Target prediction restricted to T1D risk genes was performed with miRWalk2.0. Functional annotation analysis was performed with DAVID6.7. Expression of selected microRNAs and T1D risk genes was measured by quantitative reverse-transcription polymerase chain reaction. Eighty-one microRNAs were dysregulated in persistently infected PANC-1 cells. Forty-nine of the known fifty-five T1D risk genes were predicted as putative targets of at least one of the dysregulated microRNAs. Most functional annotation terms that were enriched in these 49 putative target genes were related to the immune response or autoimmunity. mRNA levels of AFF3, BACH2, and IL7R differed significantly between persistently infected cells and uninfected cells. This is the first characterization of the microRNA expression profile changes induced by persistent coxsackievirus B4 infection in pancreatic cells. The predicted targeting of genes involved in the immune response and autoimmunity by the dysregulated microRNAs as well as the dysregulated expression of diabetes risk genes shows that persistent coxsackievirus B4 infection profoundly impacts the host cell. These data support the hypothesis of a possible link between persistent coxsackievirus B4 infection and the development of T1D.
Collapse
Affiliation(s)
- Ilka Engelmann
- Univ Lille Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, F-59000, France
| | - Enagnon K Alidjinou
- Univ Lille Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, F-59000, France
| | - Antoine Bertin
- Univ Lille Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, F-59000, France
| | - Johann Bossu
- Univ Lille Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, F-59000, France
| | - Céline Villenet
- CHU Lille, Plate-forme de Génomique Fonctionnelle et Structurale, Lille, F-59000, France
| | - Martin Figeac
- CHU Lille, Plate-forme de Génomique Fonctionnelle et Structurale, Lille, F-59000, France
| | - Famara Sane
- Univ Lille Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, F-59000, France
| | - Didier Hober
- Univ Lille Faculté de Médecine, CHU Lille, Laboratoire de Virologie EA3610, Lille, F-59000, France.
| |
Collapse
|
25
|
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: 15] [Impact Index Per Article: 1.9] [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
|
26
|
Lamborn IT, Jing H, Zhang Y, Drutman SB, Abbott JK, Munir S, Bade S, Murdock HM, Santos CP, Brock LG, Masutani E, Fordjour EY, McElwee JJ, Hughes JD, Nichols DP, Belkadi A, Oler AJ, Happel CS, Matthews HF, Abel L, Collins PL, Subbarao K, Gelfand EW, Ciancanelli MJ, Casanova JL, Su HC. Recurrent rhinovirus infections in a child with inherited MDA5 deficiency. J Exp Med 2017; 214:1949-1972. [PMID: 28606988 PMCID: PMC5502429 DOI: 10.1084/jem.20161759] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 04/13/2017] [Accepted: 05/26/2017] [Indexed: 12/15/2022] Open
Abstract
MDA5 is a cytosolic sensor of double-stranded RNA (ds)RNA including viral byproducts and intermediates. We studied a child with life-threatening, recurrent respiratory tract infections, caused by viruses including human rhinovirus (HRV), influenza virus, and respiratory syncytial virus (RSV). We identified in her a homozygous missense mutation in IFIH1 that encodes MDA5. Mutant MDA5 was expressed but did not recognize the synthetic MDA5 agonist/(ds)RNA mimic polyinosinic-polycytidylic acid. When overexpressed, mutant MDA5 failed to drive luciferase activity from the IFNB1 promoter or promoters containing ISRE or NF-κB sequence motifs. In respiratory epithelial cells or fibroblasts, wild-type but not knockdown of MDA5 restricted HRV infection while increasing IFN-stimulated gene expression and IFN-β/λ. However, wild-type MDA5 did not restrict influenza virus or RSV replication. Moreover, nasal epithelial cells from the patient, or fibroblasts gene-edited to express mutant MDA5, showed increased replication of HRV but not influenza or RSV. Thus, human MDA5 deficiency is a novel inborn error of innate and/or intrinsic immunity that causes impaired (ds)RNA sensing, reduced IFN induction, and susceptibility to the common cold virus.
Collapse
Affiliation(s)
- Ian T Lamborn
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
- Department of Pathology and Laboratory Medicine, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Huie Jing
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Yu Zhang
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Scott B Drutman
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Jordan K Abbott
- Immunodeficiency Diagnosis and Treatment Program, Division of Allergy and Immunology, Department of Pediatrics, National Jewish Health, Denver, CO
| | - Shirin Munir
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | - Heardley M Murdock
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Celia P Santos
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Linda G Brock
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Evan Masutani
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Emmanuel Y Fordjour
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | | | - Dave P Nichols
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, National Jewish Health, Denver, CO
| | - Aziz Belkadi
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR1163, Necker Hospital for Sick Children, Paris, France
- Paris Descartes University, Imagine Institute, Necker Hospital for Sick Children, Paris, France
| | - Andrew J Oler
- Bioinformatics and Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Corinne S Happel
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Helen F Matthews
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR1163, Necker Hospital for Sick Children, Paris, France
- Paris Descartes University, Imagine Institute, Necker Hospital for Sick Children, Paris, France
| | - Peter L Collins
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Kanta Subbarao
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Erwin W Gelfand
- Immunodeficiency Diagnosis and Treatment Program, Division of Allergy and Immunology, Department of Pediatrics, National Jewish Health, Denver, CO
| | - Michael J Ciancanelli
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale UMR1163, Necker Hospital for Sick Children, Paris, France
- Paris Descartes University, Imagine Institute, Necker Hospital for Sick Children, Paris, France
- Pediatric Immuno-Hematology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
- Howard Hughes Medical Institute, New York, NY
| | - Helen C Su
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
- Department of Pathology and Laboratory Medicine, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
27
|
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
|
28
|
Nonobese Diabetic (NOD) Mice Lack a Protective B-Cell Response against the "Nonlethal" Plasmodium yoelii 17XNL Malaria Protozoan. Malar Res Treat 2016; 2016:6132734. [PMID: 28074170 PMCID: PMC5198185 DOI: 10.1155/2016/6132734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/06/2016] [Accepted: 11/06/2016] [Indexed: 11/18/2022] Open
Abstract
Background. Plasmodium yoelii 17XNL is a nonlethal malaria strain in mice of different genetic backgrounds including the C57BL/6 mice (I-Ab/I-Enull) used in this study as a control strain. We have compared the trends of blood stage infection with the nonlethal murine strain of P. yoelii 17XNL malaria protozoan in immunocompetent Nonobese Diabetic (NOD) mice prone to type 1 diabetes (T1D) and C57BL/6 mice (control mice) that are not prone to T1D and self-cure the P. yoelii 17XNL infection. Prediabetic NOD mice could not mount a protective antibody response to the P. yoelii 17XNL-infected red blood cells (iRBCs), and they all succumbed shortly after infection. Our data suggest that the lack of anti-P. yoelii 17XNL-iRBCs protective antibodies in NOD mice is a result of parasite-induced, Foxp3+ T regulatory (Treg) cells able to suppress the parasite-specific antibody secretion. Conclusions. The NOD mouse model may help in identifying new mechanisms of B-cell evasion by malaria parasites. It may also serve as a more accurate tool for testing antimalaria therapeutics due to the lack of interference with a preexistent self-curing mechanism present in other mouse strains.
Collapse
|
29
|
Jaïdane H, Halouani A, Jmii H, Elmastour F, Abdelkefi S, Bodart G, Michaux H, Chakroun T, Sane F, Mokni M, Geenen V, Hober D, Aouni M. In-utero coxsackievirus B4 infection of the mouse thymus. Clin Exp Immunol 2016; 187:399-407. [PMID: 27790717 DOI: 10.1111/cei.12893] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2016] [Indexed: 12/29/2022] Open
Abstract
Type B coxsackievirus (CV-B) infections are involved frequently in the triggering of several autoimmune diseases such as myocarditis, dilated cardiomyopathy, pericarditis, pancreatitis, type 1 diabetes, encephalitis, thyroiditis or Sjögren's syndrome. Serological and virological evidence suggests that maternal infections during pregnancy can play a role in the appearance of these diseases in offspring. The current study aims to explore the effect of an in-utero CV-B infection on the fetal thymus, the central site for programming immunological self-tolerance. In this perspective, female Swiss albino mice were inoculated intraperitoneally or orally with the diabetogenic CV-B4 E2 strain at gestational days 10 or 17. Offspring were killed at different post-inoculation times, and their thymuses were analysed for evidence of infection and alterations in thymic T cell subsets. In-utero CV-B infection of the thymus was demonstrated during the course of vertical transmission, as attested by viral RNA and infectious virus detection in most analysed samples. No histopathological changes were evident. Thymic T cells were not depleted, despite being positive for viral RNA. As evidenced by flow cytometry analysis, CV-B infection of the fetal thymus induced significant changes of thymic T cell populations, particularly with maternal inoculation at gestational day 10. Altogether, these findings suggest that CV-B infection of the fetal thymus may play an important role in the genesis of autoimmune diseases.
Collapse
Affiliation(s)
- H 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
| | - A 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
| | - H Jmii
- 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
| | - F Elmastour
- 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
| | - S Abdelkefi
- Université de Sousse, Unité de recherche 'UR06SP05', Centre Régional de Transfusion Sanguine, Hôpital Farhat Hached, Sousse, Tunisia
| | - G Bodart
- Université de Liege, GIGA Research - Centre d'Immunologie, CHU-B34, B-4000 Liege-Sart, Tilman, Belgium
| | - H Michaux
- Université de Liege, GIGA Research - Centre d'Immunologie, CHU-B34, B-4000 Liege-Sart, Tilman, Belgium
| | - T Chakroun
- Université de Sousse, Unité de recherche 'UR06SP05', Centre Régional de Transfusion Sanguine, Hôpital Farhat Hached, Sousse, Tunisia
| | - F Sane
- Université Lille 2, CHRU Lille, Laboratoire de Virologie EA3610, Bâtiment P. Boulanger, Hôpital A. Calmette CHRU, Lille, 59037, France
| | - M Mokni
- Université de Sousse, CHU Farhat Hached, Service d'Anatomopathologie, Sousse, Tunisia
| | - V Geenen
- Université de Liege, GIGA Research - Centre d'Immunologie, CHU-B34, B-4000 Liege-Sart, Tilman, Belgium
| | - D Hober
- Université Lille 2, CHRU Lille, Laboratoire de Virologie EA3610, Bâtiment P. Boulanger, Hôpital A. Calmette CHRU, Lille, 59037, France
| | - M Aouni
- Université de Monastir, Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir, Monastir, Tunisia
| |
Collapse
|
30
|
Laiho JE, Oikarinen M, Richardson SJ, Frisk G, Nyalwidhe J, Burch TC, Morris MA, Oikarinen S, Pugliese A, Dotta F, Campbell-Thompson M, Nadler J, Morgan NG, Hyöty H. Relative sensitivity of immunohistochemistry, multiple reaction monitoring mass spectrometry, in situ hybridization and PCR to detect Coxsackievirus B1 in A549 cells. J Clin Virol 2016; 77:21-8. [PMID: 26875099 PMCID: PMC5364806 DOI: 10.1016/j.jcv.2016.01.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/14/2015] [Accepted: 01/28/2016] [Indexed: 11/17/2022]
Abstract
BACKGROUND Enteroviruses (EVs) have been linked to the pathogenesis of several diseases and there is a collective need to develop improved methods for the detection of these viruses in tissue samples. OBJECTIVES This study evaluates the relative sensitivity of immunohistochemistry (IHC), proteomics, in situ hybridization (ISH) and RT-PCR to detect one common EV, Coxsackievirus B1 (CVB1), in acutely infected human A549 cells in vitro. STUDY DESIGN A549 cells were infected with CVB1 and diluted with uninfected A549 cells to produce a limited dilution series in which the proportion of infected cells ranged from 10(-1) to 10(-8). Analyses were carried out by several laboratories using IHC with different anti-EV antibodies, ISH with both ViewRNA and RNAScope systems, liquid chromatography multiple reaction monitoring mass spectrometry (LC/MRM/MS/MS), and two modifications of RT-PCR. RESULTS RT-PCR was the most sensitive method for EV detection yielding positive signals in the most diluted sample (10(-8)). LC/MRM/MS/MS detected viral peptides at dilutions as high as 10(-7). The sensitivity of IHC depended on the antibody used, and the most sensitive antibody (Dako clone 5D8/1) detected virus proteins at a dilution of 10(-6), while ISH detected the virus at dilutions of 10(-4). CONCLUSIONS All methods were able to detect CVB1 in infected A549 cells. RT-PCR was most sensitive followed by LC/MRM/MS/MS and then IHC. The results from this in vitro survey suggest that all methods are suitable tools for EV detection but that their differential sensitivities need to be considered when interpreting the results from such studies.
Collapse
Affiliation(s)
- 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, Uppsala, Sweden.
| | - Julius Nyalwidhe
- Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, USA; Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, USA.
| | - Tanya C Burch
- Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, USA; Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, USA.
| | - Margaret A Morris
- Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, USA; Internal Medicine, Eastern Virginia Medical School, Norfolk, USA.
| | - Sami Oikarinen
- Department of Virology, School of Medicine, University of Tampere, Tampere, Finland.
| | - Alberto Pugliese
- Diabetes Research Institute and Departments of Medicine, Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, USA.
| | - Francesco Dotta
- Diabetes Unit, Dept. of Medicine Surgery and Neurosciences, University of Siena; Fondazione Umberto Di Mario ONLUS-Toscana Life Sciences, Siena, Italy,.
| | | | - Jerry Nadler
- Internal Medicine, Eastern Virginia Medical School, Norfolk, USA.
| | - Noel G Morgan
- University of Exeter Medical School, Exeter, Devon, UK.
| | - Heikki Hyöty
- Department of Virology, School of Medicine, University of Tampere, Tampere, Finland; Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland.
| |
Collapse
|
31
|
Elmastour F, Jaidane H, Aguech-Oueslati L, Benkahla MA, Aouni M, Gharbi J, Sane F, Hober D. Immunoglobulin G-dependent enhancement of the infection with Coxsackievirus B4 in a murine system. Virulence 2016; 7:527-35. [PMID: 27030584 DOI: 10.1080/21505594.2016.1152442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
It was demonstrated that specific IgG can enhance the infection with CV-B4, in vitro, in the human system. This enhancement could be involved in the pathophysiology of CV-B4 induced diseases. To investigate further the role of enhancing IgG in the infection with CV-B4 E2 in vivo, animal models are needed. Therefore, it was decided to assess whether inoculation of CV-B4 E2 to mice results in the appearance of IgG able to enhance the infection with this virus. Swiss albino mice were inoculated with CV-B4 E2 intraperitoneally. Serum samples were obtained from tail vein blood collected from day 0 to day 80 p.i. IgG were isolated by Protein G affinity chromatography. Seroneutralisation assays were carried out. In total murine spleen cells cultures inoculated with CV-B4 E2 mixed with various dilutions of serum or IgG samples, the enhancing activity was assayed through i) the antiviral activity titer of supernatants ii) the detection of intracellular viral RNA by RT-PCR iii) the level of infectious particles in supernatants. In most serum samples (76/105), neutralizing and enhancing activities were detected peaking between days 14 and 30 p.i and were higher in sera from mice inoculated with 2.10(6) TCID50 units than with lower doses. The enhancing activity was due to the IgG-enriched fraction of serum from CV-B4 E2 infected animals but not from control animals. These data show that IgG from immune mice can enhance the infection of splenocytes with CV-B4 E2 in vitro and open the way to explore whether such an enhancing activity can play a role in vivo.
Collapse
Affiliation(s)
- Firas Elmastour
- a Université de Lille; Faculté de Médecine, CHRU de Lille, Laboratoire de Virologie/EA3610 , Lille , France.,b Université de Monastir, Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir , Monastir , Tunisia.,c Université de Tunis El Manar, Faculté des Sciences de Tunis , Tunis , Tunisia
| | - Hela Jaidane
- b Université de Monastir, Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir , Monastir , Tunisia.,c Université de Tunis El Manar, Faculté des Sciences de Tunis , Tunis , Tunisia
| | - Leila Aguech-Oueslati
- a Université de Lille; Faculté de Médecine, CHRU de Lille, Laboratoire de Virologie/EA3610 , Lille , France.,b Université de Monastir, Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir , Monastir , Tunisia
| | - Mehdi Ayech Benkahla
- a Université de Lille; Faculté de Médecine, CHRU de Lille, Laboratoire de Virologie/EA3610 , Lille , France
| | - Mahjoub Aouni
- b Université de Monastir, Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir , Monastir , Tunisia
| | - Jawhar Gharbi
- b Université de Monastir, Laboratoire des Maladies Transmissibles et Substances Biologiquement Actives LR99ES27, Faculté de Pharmacie de Monastir , Monastir , Tunisia
| | - Famara Sane
- a Université de Lille; Faculté de Médecine, CHRU de Lille, Laboratoire de Virologie/EA3610 , Lille , France
| | - Didier Hober
- a Université de Lille; Faculté de Médecine, CHRU de Lille, Laboratoire de Virologie/EA3610 , Lille , France
| |
Collapse
|
32
|
Cabrera SM, Chen YG, Hagopian WA, Hessner MJ. Blood-based signatures in type 1 diabetes. Diabetologia 2016; 59:414-25. [PMID: 26699650 PMCID: PMC4744128 DOI: 10.1007/s00125-015-3843-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/18/2015] [Indexed: 12/15/2022]
Abstract
Type 1 diabetes mellitus is one of the most common chronic diseases in childhood. It develops through autoimmune destruction of the pancreatic beta cells and results in lifelong dependence on exogenous insulin. The pathogenesis of type 1 diabetes involves a complex interplay of genetic and environmental factors and has historically been attributed to aberrant adaptive immunity; however, there is increasing evidence for a role of innate inflammation. Over the past decade new methodologies for the analysis of nucleic acid and protein signals have been applied to type 1 diabetes. These studies are providing a new understanding of type 1 diabetes pathogenesis and have the potential to inform the development of new biomarkers for predicting diabetes onset and monitoring therapeutic interventions. In this review we will focus on blood-based signatures in type 1 diabetes, with special attention to both direct transcriptomic analyses of whole blood and immunocyte subsets, as well as plasma/serum-induced transcriptional signatures. Attention will also be given to proteomics, microRNA assays and markers of beta cell death. We will also discuss the results of blood-based profiling in type 1 diabetes within the context of the genetic and environmental factors implicated in the natural history of autoimmune diabetes.
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, Milwaukee, WI, USA
- Section of Endocrinology, Department of Pediatrics, The Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Yi-Guang Chen
- The Max McGee National Research Center for Juvenile Diabetes, Children's Research Institute of Children's Hospital of Wisconsin, Milwaukee, WI, USA
- Section of Endocrinology, Department of Pediatrics, 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, Milwaukee, WI, USA.
- Section of Endocrinology, Department of Pediatrics, The Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
33
|
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
|
34
|
Michaux H, Martens H, Jaïdane H, Halouani A, Hober D, Geenen V. How Does Thymus Infection by Coxsackievirus Contribute to the Pathogenesis of Type 1 Diabetes? Front Immunol 2015; 6:338. [PMID: 26175734 PMCID: PMC4485212 DOI: 10.3389/fimmu.2015.00338] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/17/2015] [Indexed: 12/16/2022] Open
Abstract
Through synthesis and presentation of neuroendocrine self-antigens by major histocompatibility complex proteins, thymic epithelial cells (TECs) play a crucial role in programing central immune self-tolerance to neuroendocrine functions. Insulin-like growth factor-2 (IGF-2) is the dominant gene/polypeptide of the insulin family that is expressed in TECs from different animal species and humans. Igf2 transcription is defective in the thymus of diabetes-prone bio-breeding rats, and tolerance to insulin is severely decreased in Igf2 (-/-) mice. For more than 15 years now, our group is investigating the hypothesis that, besides a pancreotropic action, infection by coxsackievirus B4 (CV-B4) could implicate the thymus as well, and interfere with the intrathymic programing of central tolerance to the insulin family and secondarily to insulin-secreting islet β cells. In this perspective, we have demonstrated that a productive infection of the thymus occurs after oral CV-B4 inoculation of mice. Moreover, our most recent data have demonstrated that CV-B4 infection of a murine medullary (m) TEC line induces a significant decrease in Igf2 expression and IGF-2 production. In these conditions, Igf1 expression was much less affected by CV-B4 infection, while Ins2 transcription was not detected in this cell line. Through the inhibition of Igf2 expression in TECs, CV-B4 infection could lead to a breakdown of central immune tolerance to the insulin family and promote an autoimmune response against insulin-secreting islet β cells. Our major research objective now is to understand the molecular mechanisms by which CV-B4 infection of TECs leads to a major decrease in Igf2 expression in these cells.
Collapse
Affiliation(s)
- Hélène Michaux
- Department of Biomedical and Preclinical Sciences, GIGA-I 3 Center of Immunoendocrinology, GIGA Research Institute, University of Liege , Liege , Belgium
| | - Henri Martens
- Department of Biomedical and Preclinical Sciences, GIGA-I 3 Center of Immunoendocrinology, GIGA Research Institute, University of Liege , Liege , Belgium
| | - Hela Jaïdane
- Laboratory of Virology LR99ES27, School of Pharmacy, University of Monastir , Monastir , Tunisia ; Faculty of Sciences of Tunis, University of Tunis El Manar , Tunis , Tunisia
| | - Aymen Halouani
- Laboratory of Virology LR99ES27, School of Pharmacy, University of Monastir , Monastir , Tunisia ; Faculty of Sciences of Tunis, University of Tunis El Manar , Tunis , Tunisia
| | - Didier Hober
- Laboratory of Virology EA3610, Centre Hospitalier Régional Universitaire de Lille, University of Lille 2 , Lille , France
| | - Vincent Geenen
- Department of Biomedical and Preclinical Sciences, GIGA-I 3 Center of Immunoendocrinology, GIGA Research Institute, University of Liege , Liege , Belgium
| |
Collapse
|
35
|
Bergamin CS, Dib SA. Enterovirus and type 1 diabetes: What is the matter? World J Diabetes 2015; 6:828-839. [PMID: 26131324 PMCID: PMC4478578 DOI: 10.4239/wjd.v6.i6.828] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/30/2015] [Accepted: 04/09/2015] [Indexed: 02/05/2023] Open
Abstract
A complex interaction of genetic and environmental factors can trigger the immune-mediated mechanism responsible for type 1 diabetes mellitus (T1DM) establishment. Environmental factors may initiate and possibly sustain, accelerate, or retard damage to β-cells. The role of environmental factors in this process has been exhaustive studied and viruses are among the most probable ones, especially enteroviruses. Improvements in enterovirus detection methods and randomized studies with patient follow-up have confirmed the importance of human enterovirus in the pathogenesis of T1DM. The genetic risk of T1DM and particular innate and acquired immune responses to enterovirus infection contribute to a tolerance to T1DM-related autoantigens. However, the frequency, mechanisms, and pathways of virally induced autoimmunity and β-cell destruction in T1DM remain to be determined. It is difficult to investigate the role of enterovirus infection in T1DM because of several concomitant mechanisms by which the virus damages pancreatic β-cells, which, consequently, may lead to T1DM establishment. Advances in molecular and genomic studies may facilitate the identification of pathways at earlier stages of autoimmunity when preventive and therapeutic approaches may be more effective.
Collapse
|
36
|
Application of bioinformatics in probe design enables detection of enteroviruses on different taxonomic levels by advanced in situ hybridization technology. J Clin Virol 2015. [PMID: 26209400 DOI: 10.1016/j.jcv.2015.06.085] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND Enteroviral infections are common, affecting humans across all age groups. RT-PCR is widely used to detect these viruses in clinical samples. However, there is a need for sensitive and specific in situ detection methods for formalin-fixed tissues, allowing for the anatomical localization of the virus and identification of its serotype. OBJECTIVES The aim was to design novel enterovirus probes, assess the impact of probe design for the detection and optimize the new single molecule in situ hybridization technology for the detection of enteroviruses in formalin-fixed paraffin-embedded samples. STUDY DESIGN Four enterovirus RNA-targeted oligonucleotide RNA probes - two probes for wide range enterovirus detection and two for serotype-targeted detection of Coxsackievirus B1 (CVB1) - were designed and validated for the commercially available QuantiGene ViewRNA in situ hybridization method. The probe specificities were tested using a panel of cell lines infected with different enterovirus serotypes and CVB infected mouse pancreata. RESULTS The two widely reactive probe sets recognized 19 and 20 of the 20 enterovirus serotypes tested, as well as 27 and 31 of the 31 CVB1 strains tested. The two CVB1 specific probe sets detected 30 and 14 of the 31 CVB1 strains, with only minor cross-reactivity to other serotypes. Similar results were observed in stained tissues from CVB -infected mice. CONCLUSIONS These novel in-house designed probe sets enable the detection of enteroviruses from formalin-fixed tissue samples. Optimization of probe sequences makes it possible to tailor the assay for the detection of enteroviruses on the serotype or species level.
Collapse
|
37
|
Alidjinou EK, Sané F, Bertin A, Caloone D, Hober D. Persistent infection of human pancreatic cells with Coxsackievirus B4 is cured by fluoxetine. Antiviral Res 2015; 116:51-4. [PMID: 25655448 DOI: 10.1016/j.antiviral.2015.01.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 01/12/2015] [Accepted: 01/25/2015] [Indexed: 12/16/2022]
Abstract
Group B Coxsackieviruses (CVB) are involved in various acute clinical features and they can play a role in the development of chronic diseases like type 1 diabetes. The persistence of CVB has been described in vitro and in vivo in various models. Fluoxetine was reported to inhibit the replication of CVB1-3, which prompted us to study the in vitro antiviral activity of fluoxetine against CVB4 in models of acute infection. In addition we took advantage of a chronically CVB4-infected Panc-1 cell line to evaluate the antiviral effect of fluoxetine in a model of persistent CVB4 infection. An inhibition of the CVB4 replication was obtained when fluoxetine was added at 5.48μM to Hep-2 cell cultures. No inhibitory effect was observed when CVB4 was mixed with fluoxetine for 2h and filtered to eliminate fluoxetine before inoculation to cells, or when cells were treated up to 96h and washed before viral inoculation. Fluoxetine (5.48μM) reduced viral replication by more than 50% in acutely infected Panc-1 cell cultures. A dramatic decrease of infectious particles levels in supernatants of Panc-1 cells chronically infected with CVB4 was obtained a few days after treatment with fluoxetine and no infectious viral particle was found as soon as day 21 of treatment, and intracellular enteroviral RNA was undetectable by RT-PCR after three weeks of treatment. These data display that fluoxetine can inhibit the replication of CVB4 and can cure Panc-1 cells chronically infected with CVB4.
Collapse
Affiliation(s)
- Enagnon Kazali Alidjinou
- Université Lille 2, Faculté de Médecine, CHRU de Lille, Laboratoire de virologie EA3610, Lille, France
| | - Famara Sané
- Université Lille 2, Faculté de Médecine, CHRU de Lille, Laboratoire de virologie EA3610, Lille, France
| | - Antoine Bertin
- Université Lille 2, Faculté de Médecine, CHRU de Lille, Laboratoire de virologie EA3610, Lille, France
| | - Delphine Caloone
- Université Lille 2, Faculté de Médecine, CHRU de Lille, Laboratoire de virologie EA3610, Lille, France
| | - Didier Hober
- Université Lille 2, Faculté de Médecine, CHRU de Lille, Laboratoire de virologie EA3610, Lille, France.
| |
Collapse
|
38
|
Pompei R. The Role of Human Herpesvirus 8 in Diabetes Mellitus Type 2: State of the Art and a Medical Hypothesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 901:37-45. [PMID: 26542602 DOI: 10.1007/5584_2015_5014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Diabetes is a common chronic disease due to an altered glucose metabolism, caused by the quantitative and/or qualitative dysfunction of the insulin hormone. Two types of diabetes are recognized: juvenile diabetes, or type 1, which has an autoimmune origin, and adult diabetes, or type 2 (DMT2), which covers 90-95 % of all diabetic patients.The causes of DMT2 are not yet clear: heredity, life style, nutrition, and environment are considered the main risk factors. Several viral infections, namely cytomegalovirus, coxsackie and other enteroviruses, rubella and hepatitis C virus, have been claimed to be associated with some forms of diabetes. The direct role of viruses as a cause or as a risk of type 1 diabetes has been amply described in several recent reviews. Therefore, this review focuses attention on the role of a human herpes pathogenic virus in the onset of DMT2. By carrying out an analysis of recent literature, we describe the findings reported on an extremely deceitful virus, such as Human Herpes virus 8, and present a medical hypothesis on a possible relationship between this virus and DMT2.
Collapse
Affiliation(s)
- Raffaello Pompei
- Department of Biomedical Sciences, University of Cagliari, via Porcell 4, 09124, Cagliari, Italy.
| |
Collapse
|
39
|
Emerging and Reemerging Infectious Disease Threats. MANDELL, DOUGLAS, AND BENNETT'S PRINCIPLES AND PRACTICE OF INFECTIOUS DISEASES 2015. [PMCID: PMC7151803 DOI: 10.1016/b978-1-4557-4801-3.00014-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
40
|
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
|
41
|
Adamczak DM, Nowak JK, Frydrychowicz M, Kaczmarek M, Sikora J. The role of Toll-like receptors and vitamin D in diabetes mellitus type 1--a review. Scand J Immunol 2014; 80:75-84. [PMID: 24845558 DOI: 10.1111/sji.12188] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/04/2014] [Indexed: 12/17/2022]
Abstract
It is widely accepted that type 1 diabetes mellitus (T1DM) is an autoimmune disease resulting from an interaction between immunologic, genetic and environmental factors. However, the exact mechanism leading to the development of T1DM remains incomplete. There is a large body of evidence pointing towards the important role of toll-like receptor (TLR) activation and vitamin D deficiency in T1DM pathogenesis. In this article, we review the available data on the influence of TLRs' level of activation and vitamin D status on the risk of the development of T1DM in humans and rodent models. We also summarize the current information regarding the interactions between TLRs' level of activation, vitamin D status and various environmental factors, such as enteroviral infections, the gut microbiota and breastfeeding substitution, among others. Our results stipulate that vitamin D seems to protect against T1DM by reducing the TLRs' level of activation.
Collapse
Affiliation(s)
- D M Adamczak
- Poznan University of Medical Sciences, Clinical Hospital No. 1, Poznan, Poland; Department of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | | | | |
Collapse
|
42
|
Liu WY, Lu DJ, Du XM, Sun JQ, Ge J, Wang RW, Wang R, Zou J, Xu C, Ren J, Wen XF, Liu Y, Cheng SM, Tan X, Pekkala S, Munukka E, Wiklund P, Chen YQ, Gu Q, Xia ZC, Liu JJ, Liu WB, Chen XB, Zhang YM, Li R, Borra RJH, Yao JX, Chen PJ, Cheng S. Effect of aerobic exercise and low carbohydrate diet on pre-diabetic non-alcoholic fatty liver disease in postmenopausal women and middle aged men--the role of gut microbiota composition: study protocol for the AELC randomized controlled trial. BMC Public Health 2014; 14:48. [PMID: 24438438 PMCID: PMC3897962 DOI: 10.1186/1471-2458-14-48] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 01/14/2014] [Indexed: 02/07/2023] Open
Abstract
Background Pre-diabetes and non-alcoholic fatty liver disease (NAFLD) are associated with an unhealthy lifestyle and pose extremely high costs to the healthcare system. In this study, we aim to explore whether individualized aerobic exercise (AEx) and low carbohydrate diet (LCh) intervention affect hepatic fat content (HFC) in pre-diabetes via modification of gut microbiota composition and other post-interventional effects. Methods/design A 6-month randomized intervention with 6-month follow-up is conducted from January 2013 to December 2015. The target sample size for intervention is 200 postmenopausal women and middle-aged men aged 50–65 year-old with pre-diabetes and NAFLD. The qualified subjects are randomized into 4 groups with 50 subjects in each group: 1 = AEx, 2 = LCh, 3 = AEx + LCh, and 4 = control. In addition, two age-matched reference groups (5 = pre-diabetes without NAFLD (n = 50) and 6 = Healthy without pre-diabetes or NAFLD (n = 50)) are included. The exercise program consists of progressive and variable aerobic exercise (intensity of 60 to 75% of initial fitness level, 3–5 times/week and 30–60 min/time). The diet program includes dietary consultation plus supplementation with a special lunch meal (40% of total energy intake/day) which aims to reduce the amount of carbohydrate consumption (30%). The control and reference groups are advised to maintain their habitual habits during the intervention. The primary outcome measures are HFC, serum metabolomics and gut microbiota composition. The secondary outcome measures include body composition and cytokines. In addition, socio-psychological aspects, social support, physical activity and diet will be performed by means of questionnaire and interview. Discussion Specific individualized exercise and diet intervention in this study offers a more efficient approach for liver fat reduction and diabetes prevention via modification of gut microbiota composition. Besides, the study explores the importance of incorporating fitness assessment and exercise in the management of patients with pre-diabetes and fatty liver disorders. If our program is shown to be effective, it will open new strategies to combat these chronic diseases. Trial registration Current Controlled Trials: ISRCTN42622771.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Pei Jie Chen
- School of Kinesiology, Shanghai University of Sport, 200438 Shanghai, China.
| | | |
Collapse
|
43
|
The A allele of the rs1990760 polymorphism in the IFIH1 gene is associated with protection for arterial hypertension in type 1 diabetic patients and with expression of this gene in human mononuclear cells. PLoS One 2013; 8:e83451. [PMID: 24386202 PMCID: PMC3873949 DOI: 10.1371/journal.pone.0083451] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 11/04/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The rs1990760 polymorphism of interferon induced with helicase C domain 1 (IFIH1) has been associated with type 1 diabetes mellitus (T1DM). Here, we investigated whether this polymorphism is associated with T1DM or its clinical characteristics in a Brazilian population, and if IFIH1 gene expression in mononuclear cells from T1DM patients differs according to the genotypes of this polymorphism. A meta-analysis was also conducted to evaluate if the rs1990760 polymorphism is associated with T1DM. METHODS Frequencies of the rs1990760 polymorphism were analyzed in 527 T1DM patients and in 517 healthy subjects. IFIH1 gene expressions according to genotypes were measured in a sub-sample of 26 T1DM patients by quantitative real-time PCR. RESULTS Our data show the association of the A allele with risk to T1DM under a dominant model of inheritance [odds ratio (OR) = 1.421, P = 0.037], adjusting for ethnicity. The meta-analysis revealed significant association between the rs199760A allele and risk for T1DM for all analyzed inheritance models. Surprisingly, T1DM patients carrying the A allele showed lower levels of systolic (P = 0.001) and diastolic (P = 1 × 10(-10)) blood pressures as compared to G/G carriers. Furthermore, the A/A genotype seems to be associated with protection to arterial hypertension (AH) after adjustment for covariates (OR = 0.339, P = 0.019). IFIH1 gene expression in mononuclear cells from 26 T1DM patients did not differ among genotypes (P = 0.274). Nevertheless, IFIH1 gene expression was increased in mononuclear cells from T1DM patients with AH as compared with T1DM patients without AH [6.7 (1.7-2.0) vs. 1.8 (1.3-7.1) arbitrary units; P = 0.036]. The association with blood pressures and AH was not observed in patients with type 2 diabetes mellitus. CONCLUSIONS Our results indicate that the rs1990760 polymorphism is associated with T1DM. Interestingly, the rs1990760 A allele seems to be associated with protection for AH in T1DM patients. Further studies are needed to confirm the association with AH.
Collapse
|
44
|
Serum-Dependent Enhancement of Coxsackievirus B4-Induced Production of IFNα, IL-6 and TNFα by Peripheral Blood Mononuclear Cells. J Mol Biol 2013; 425:5020-31. [DOI: 10.1016/j.jmb.2013.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 09/24/2013] [Accepted: 10/03/2013] [Indexed: 12/21/2022]
|
45
|
Sane F, Caloone D, Gmyr V, Engelmann I, Belaich S, Kerr-Conte J, Pattou F, Desailloud R, Hober D. Coxsackievirus B4 can infect human pancreas ductal cells and persist in ductal-like cell cultures which results in inhibition of Pdx1 expression and disturbed formation of islet-like cell aggregates. Cell Mol Life Sci 2013; 70:4169-80. [PMID: 23775130 PMCID: PMC11113870 DOI: 10.1007/s00018-013-1383-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 05/14/2013] [Accepted: 05/23/2013] [Indexed: 12/31/2022]
Abstract
The role of enteroviruses, especially Coxsackievirus B (CVB), in type 1 diabetes is suspected, but the mechanisms of the virus-induced or aggravated pathogenesis of the disease are unknown. The hypothesis of an enterovirus-induced disturbance of pancreatic β-cells regeneration has been investigated in the human system. The infection of human pancreas ductal cells and pancreatic duct cell line, PANC-1, with CVB4E2 has been studied. Primary ductal cells and PANC-1 cells were infectable with CVB4E2 and a RT-PCR assay without extraction displayed that a larger proportion of cells harbored viral RNA than predicted by the detection of the viral capsid protein VP1 by indirect immunofluorescence. The detection of intracellular positive- and negative-strands of enterovirus genomes in cellular extracts by RT-PCR and the presence of infectious particles in supernatant fluids during the 37 weeks of monitoring demonstrated that CVB4E2 could persist in the pancreatic duct cell line. A persistent infection of these cells resulted in an impaired expression of Pdx1, a transcription factor required for the formation of endocrine pancreas, and a disturbed formation of islet-like cell aggregates of which the viability was decreased. These data support the hypothesis of an impact of enteroviruses onto pancreatic ductal cells which are involved in the renewal of pancreatic β-cells.
Collapse
Affiliation(s)
- Famara Sane
- Laboratoire de Virologie/ EA3610, Université Lille 2, Faculté de Médecine, CHRU, 59120 Loos-lez-Lille, France
| | - Delphine Caloone
- Laboratoire de Virologie/ EA3610, Université Lille 2, Faculté de Médecine, CHRU, 59120 Loos-lez-Lille, France
| | - Valéry Gmyr
- Laboratoire Biothérapie du diabète, INSERM U859 CHRU de Lille, 59045, Lille, France
| | - Ilka Engelmann
- Laboratoire de Virologie/ EA3610, Université Lille 2, Faculté de Médecine, CHRU, 59120 Loos-lez-Lille, France
| | - Sandrine Belaich
- Laboratoire Biothérapie du diabète, INSERM U859 CHRU de Lille, 59045, Lille, France
| | - Julie Kerr-Conte
- Laboratoire Biothérapie du diabète, INSERM U859 CHRU de Lille, 59045, Lille, France
| | - François Pattou
- Laboratoire Biothérapie du diabète, INSERM U859 CHRU de Lille, 59045, Lille, France
| | - Rachel Desailloud
- Service d’Endocrinologie-Diabétologie-Nutrition, UPJV CHU, 80054 Amiens, France
| | - Didier Hober
- Laboratoire de Virologie/ EA3610, Université Lille 2, Faculté de Médecine, CHRU, 59120 Loos-lez-Lille, France
- Laboratoire de Virologie/EA3610, Institut Hippocrate, CHRU Lille, 152 rue du Dr Yersin, 59120 Loos-Lez-Lille, France
| |
Collapse
|
46
|
Pothlichet J, Quintana-Murci L. The genetics of innate immunity sensors and human disease. Int Rev Immunol 2013; 32:157-208. [PMID: 23570315 DOI: 10.3109/08830185.2013.777064] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Since their discovery, innate immunity microbial sensors have been increasingly studied and shown to play a critical role in innate responses to microbes in several experimental in vitro, ex vivo, and animal models. However, their role in the human response to infection in natural conditions has just started to be deciphered, by means of clinical studies of primary immunodeficiencies and epidemiological genetic studies. Here, we summarize the major findings concerning the genetic diversity of the various families of microbial sensors in humans, and of other molecules involved in the signaling pathways they trigger. Specifically, we review the genetic associations, revealed by both clinical and epidemiological genetics studies, of microbial sensors from five different families: Toll-like receptors, C-type lectin receptors, NOD-like receptors, RIG-I-like receptors, and cytosolic DNA sensors. In particular, we consider the relationships between variation at the genes encoding these molecules and susceptibility to and the severity of infectious diseases and other clinical conditions associated with immune dysfunction, including autoimmunity, inflammation, allergy, and cancer. Despite the fact that the genetic links between innate immunity sensors and human disorders remain still limited, human genetics studies are increasingly improving our understanding of the genuine functions of microbial sensors and downstream signaling molecules in the natural setting.
Collapse
Affiliation(s)
- Julien Pothlichet
- Institut Pasteur, Unit of Human Evolutionary Genetics, Paris, France
| | | |
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW Type 1 diabetes (T1D) results from interplay between genetic predisposition, immune system, and environmental factors. Epidemiological and experimental data strongly suggest a role for enteroviruses in the development of T1D, but a lot of controversies and unanswered questions remained. This review focuses on issues that are fueling debate. RECENT FINDINGS Beyond HLA genes, which provide genetic susceptibility for T1D, other loci have been identified to be associated with the disease. There is a link between T1D and single-nucleotide polymorphisms (SNPs) in the interferon-induced helicase 1 (IFIH1) gene that encodes melanoma differentiation-associated protein 5 (MDA5). This protein is a cytoplasmic sensor for viruses especially coxsackieviruses B, the most incriminated enteroviruses in T1D pathogenesis. Upon viral infection, MDA5 stimulates the production of mediators of the innate antiviral immune response, which is believed to play a role in a 'bystander activation' scenario. Rare variants of IFIH1 through a lost or reduced expression of the protein are protective against T1D, whereas common IFIH1 SNPs are associated with the disease. However, a clear association has not been yet established between T1D-associated IFIH1 polymorphisms and enterovirus detection. SUMMARY Literature have accumulated a lot of evidence supporting that enteroviruses can contribute, at least in some patients, to the pathogenesis of T1D through various mechanisms. But it is still a challenge to date to prove a causal relationship between enteroviruses and T1D. Future studies may lead to a better understanding of this relationship and ultimately can help toward disease prevention.
Collapse
|
48
|
Kaçar N, Cevahir N, Demirkan N, Şanlı B. The investigation of the possible relationship between Coxsackie viruses and pemphigus. Int J Dermatol 2013; 53:312-5. [DOI: 10.1111/j.1365-4632.2012.05699.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Nida Kaçar
- Departments of Dermatology, Microbiology, and Pathology; Pamukkale University; Denizli Turkey
| | - Nural Cevahir
- Departments of Dermatology, Microbiology, and Pathology; Pamukkale University; Denizli Turkey
| | - Neşe Demirkan
- Departments of Dermatology, Microbiology, and Pathology; Pamukkale University; Denizli Turkey
| | - Berna Şanlı
- Departments of Dermatology, Microbiology, and Pathology; Pamukkale University; Denizli Turkey
| |
Collapse
|
49
|
Harris KG, Coyne CB. Enter at your own risk: how enteroviruses navigate the dangerous world of pattern recognition receptor signaling. Cytokine 2013; 63:230-6. [PMID: 23764548 DOI: 10.1016/j.cyto.2013.05.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 05/09/2013] [Indexed: 12/25/2022]
Abstract
Enteroviruses are the most common human viral pathogens worldwide. This genus of small, non-enveloped, single stranded RNA viruses includes coxsackievirus, rhinovirus, echovirus, and poliovirus species. Infection with these viruses can induce mild symptoms that resemble the common cold, but can also be associated with more severe syndromes such as poliomyelitis, neurological diseases including aseptic meningitis and encephalitis, myocarditis, and the onset of type I diabetes. In humans, polarized epithelial cells lining the respiratory and/or digestive tracts represent the initial sites of infection by enteroviruses. Control of infection in the host is initiated through the engagement of a variety of pattern recognition receptors (PRRs). PRRs act as the sentinels of the innate immune system and serve to alert the host to the presence of a viral invader. This review assembles the available data annotating the role of PRRs in the response to enteroviral infection as well as the myriad ways by which enteroviruses both interrupt and manipulate PRR signaling to enhance their own replication, thereby inducing human disease.
Collapse
Affiliation(s)
- Katharine G Harris
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 427 Bridgeside Point II, 450 Technology Drive, Pittsburgh, PA 15219, USA.
| | | |
Collapse
|
50
|
Cubas-Dueñas I, Cabrera-Rode E, Sarmiento L, Molina G, Fonseca M, Arranz C, Domínguez E, González P, Vera M, Díaz-Horta O. First-degree relatives of persons with type 1 diabetes: insulin resistance and enterovirus infection are associated with different patterns of islet cell autoimmunity. Acta Diabetol 2013; 50:233-9. [PMID: 21604002 DOI: 10.1007/s00592-011-0297-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 05/05/2011] [Indexed: 01/27/2023]
Abstract
Type 1 diabetes (T1D) results from the interaction of genetic and environmental factors. Previous studies indicate an association between detection of Enterovirus (EV) genome in blood and the clinical onset of T1D. Insulin resistance can also represent a risk factor for progression to clinically overt T1D. This study aimed at evaluating whether there is association between both EV infection and insulin resistance with islet autoantibodies in first-degree relatives of persons with type 1 diabetes. We collected sera from 94 first-degree relatives with (32) or without (64) islet cell antibodies (ICA) from the Cuban T1D prediction program. Blood glucose and insulin concentrations were determined. Antibodies to GAD65 and IA-2 were determined by radioimmunoassay. Insulin resistance was estimated by the homeostasis model assessment (HOMA-IR). EV-RNA was detected in serum using a highly sensitive reverse transcriptase-polymerase chain reaction method. The occurrence of EV-RNA was higher in ICA-positive relatives than in ICA-negative ones [15.6% (5/32) vs. 1.6% (1/62), P = 0.016]. GAD65 autoantibodies were more frequent in subjects with insulin resistance [34.5% (10/29) vs. 13.9% (9/65), P = 0.028] as defined by the HOMA-IR value. GAD65 autoantibodies also positively correlated with HOMA-IR (r.bis = 0.28, P < 0.01). IA-2 autoantibodies did correlate neither with EV-RNA nor with insulin resistance. There was no association between the presence of EV-RNA and insulin resistance. Our data suggest that enterovirus infection and insulin resistance are two independent events associated with ICA and GAD65 autoantibodies, respectively. These observations support the multifactorial nature of T1D.
Collapse
Affiliation(s)
- Ileana Cubas-Dueñas
- Department of Immunology and Genetics on Diabetes, National Institute of Endocrinology, Zapata and D, 10400, Havana, Cuba
| | | | | | | | | | | | | | | | | | | |
Collapse
|