1
|
Tzang CC, Chi LY, Lee CY, Chang ZY, Luo CA, Chen YH, Lin TA, Yu LC, Chen YR, Tzang BS, Hsu TC. Clinical implications of human Parvovirus B19 infection on autoimmunity and autoimmune diseases. Int Immunopharmacol 2025; 147:113960. [PMID: 39746271 DOI: 10.1016/j.intimp.2024.113960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/05/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025]
Abstract
Parvovirus B19 (B19V) is a human pathogen from the Parvoviridae family that primarily targets and replicates in erythroid progenitor cells (EPCs). While its symptoms are typically self-limiting in healthy individuals, B19V can cause or exacerbate autoimmune diseases in vulnerable patients. This review integrates the involvement of B19V in the development and worsening of several autoimmune diseases, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), juvenile idiopathic arthritis (JIA), hematological disorders (thalassemia, anemia, and thrombocytopenia), vasculitis, antiphospholipid syndrome (APS), dermatological disease (systemic sclerosis, psoriasis), autoimmune thyroid disease, myocarditis, and myasthenia gravis, and autoinflammatory disease of adult-onset Still's disease (AOSD). B19V contributes to autoimmunity and autoimmune disease onset and progression through mechanisms such as molecular mimicry, immune system disruption, and chronic infection. By summarizing findings from in vitro experiments, clinical case studies, seroprevalence data, and biopsy results, this review highlights the critical connection between B19V and autoimmune disease development. Recognizing the role of B19V in the early diagnosis and management of these conditions is essential, as its presence may influence the disease course and severity. Greater awareness among healthcare professionals and the public is necessary to address the impact of B19V, leading to more accurate diagnoses and better-informed treatment approaches for autoimmune diseases linked to the virus.
Collapse
Affiliation(s)
- Chih-Chen Tzang
- School of Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Liang-Yun Chi
- School of Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chen-Yu Lee
- School of Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Zi-Yi Chang
- School of Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chiao-An Luo
- School of Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yan-Hua Chen
- School of Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Tzu-An Lin
- School of Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Liang-Chien Yu
- School of Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yo-Rong Chen
- School of Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Bor-Show Tzang
- Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan; Immunology Research Center, Chung Shan Medical University, Taichung 402, Taiwan.
| | - Tsai-Ching Hsu
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan; Immunology Research Center, Chung Shan Medical University, Taichung 402, Taiwan.
| |
Collapse
|
2
|
Guo M, Shang S, Li M, Cai G, Li P, Chen X, Li Q. Understanding autoimmune response after SARS-CoV-2 infection and the pathogenesis/mechanisms of long COVID. MEDICAL REVIEW (2021) 2024; 4:367-383. [PMID: 39444797 PMCID: PMC11495526 DOI: 10.1515/mr-2024-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/04/2024] [Indexed: 10/25/2024]
Abstract
COVID-19 posed a major challenge to the healthcare system and resources worldwide. The popularization of vaccines and the adoption of numerous prevention and control measures enabled the gradual end of the COVID-19 pandemic. However, successive occurrence of autoimmune diseases in patients with COVID-19 cannot be overlooked. Long COVID has been the major focus of research due to the long duration of different symptoms and the variety of systems involved. Autoimmunity may play a crucial role in the pathogenesis of long COVID. Here, we reviewed several autoimmune disorders occurring after COVID-19 infection and the pathogenesis of long COVID.
Collapse
Affiliation(s)
- Ming Guo
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Shunlai Shang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Mengfei Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
- Haihe Laboratory of CellEcosystem, China
| | - Qinggang Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| |
Collapse
|
3
|
Allen EK, Penkert RR, Hankins JS, Surman SL, Van de Velde LA, Cotton A, Hayden RT, Tang L, Yuan X, Zheng Y, Thomas PG, Hurwitz JL. Immune Cell Profiles of Patients with Sickle Cell Disease during Parvovirus B19-Induced Transient Red Cell Aplasia. Vaccines (Basel) 2024; 12:984. [PMID: 39340016 PMCID: PMC11435734 DOI: 10.3390/vaccines12090984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/11/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
Parvovirus B19 frequently infects children and targets cells of the erythroid lineage. Although healthy children rarely suffer severe disease, children with sickle cell disease (SCD) can experience transient red cell aplasia (TRCA), hospitalization, and life-threatening anemia upon first virus exposure. Given that children with SCD can also suffer chronic inflammation and that parvovirus B19 has been associated with autoimmune disease in other patient populations, we asked if parvovirus B19 infections contributed to acute and chronic immune abnormalities in children with SCD. Nineteen hospitalized patients with SCD and parvovirus B19-induced TRCA were evaluated. Blood tests included CBC, flow cytometry, and total antibody isotype analyses. Cytokine/chemokine analyses were performed on nasal wash (NW) samples, representing a common site of viral entry. Unusually high white blood cell count (WBC) and absolute neutrophil count (ANC) values were observed in some patients. A correlation matrix with Day 0 values from the 19 patients then identified two mutually exclusive phenotype clusters. Cluster 1 included WBC, ANC, absolute reticulocyte count (ARC), absolute lymphocyte count (ALC), lactate dehydrogenase (LDH), NW cytokines/chemokines, % naïve cells among B cell and T cell populations, and parvovirus-specific IgG. This cluster was negatively associated with virus load, suggesting a signature of successful adaptive immunity and virus control. Cluster 2 included virus load, % CD38+CD24- cells among CD19+ B cells (termed 'plasmablasts' for simplicity), % HLA-DRlow cells among CD19+ B cells, IgG4, and % memory phenotypes among B cell and T cell populations. Plasmablast percentages correlated negatively with parvovirus-specific IgG, possibly reflecting a non-specific trigger of cell activation. All patients were released from the hospital within 1 week after admission, and the highest WBC and ANC values were eventually reduced. Nonetheless, a concern remained that the acutely abnormal immune profiles caused by parvovirus B19 infections could exacerbate chronic inflammation in some patients. To avoid the numerous sequelae known to affect patients with SCD following hospitalizations with parvovirus B19, rapid development of a parvovirus B19 vaccine is warranted.
Collapse
Affiliation(s)
- E. Kaitlynn Allen
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (E.K.A.); (L.-A.V.d.V.); (P.G.T.)
| | - Rhiannon R. Penkert
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.R.P.); (S.L.S.)
| | - Jane S. Hankins
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.S.H.); (A.C.)
- Department of Global Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Sherri L. Surman
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.R.P.); (S.L.S.)
| | - Lee-Ann Van de Velde
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (E.K.A.); (L.-A.V.d.V.); (P.G.T.)
| | - Alyssa Cotton
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.S.H.); (A.C.)
| | - Randall T. Hayden
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Li Tang
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (L.T.); (X.Y.); (Y.Z.)
| | - Xiaomeng Yuan
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (L.T.); (X.Y.); (Y.Z.)
| | - Ying Zheng
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (L.T.); (X.Y.); (Y.Z.)
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (E.K.A.); (L.-A.V.d.V.); (P.G.T.)
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Julia L. Hurwitz
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (R.R.P.); (S.L.S.)
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
4
|
Arvia R, Stincarelli MA, Manaresi E, Gallinella G, Zakrzewska K. Parvovirus B19 in Rheumatic Diseases. Microorganisms 2024; 12:1708. [PMID: 39203550 PMCID: PMC11357344 DOI: 10.3390/microorganisms12081708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/03/2024] Open
Abstract
Parvovirus B19 (B19V) is a human pathogen belonging to the Parvoviridae family. It is widely diffused in the population and responsible for a wide range of diseases, diverse in pathogenetic mechanisms, clinical course, and severity. B19V infects and replicates in erythroid progenitor cells (EPCs) in the bone marrow leading to their apoptosis. Moreover, it can also infect, in an abortive manner, a wide set of different cell types, normally non-permissive, and modify their normal physiology. Differences in the characteristics of virus-cell interaction may translate into different pathogenetic mechanisms and clinical outcomes. Joint involvement is a typical manifestation of B19V infection in adults. Moreover, several reports suggest, that B19V could be involved in the pathogenesis of some autoimmune rheumatologic diseases such as rheumatoid arthritis (RA), juvenile idiopathic arthritis (JIA), systemic sclerosis (SSc), systemic lupus erythematosus (SLE), or vasculitis. This review provides basic information on the B19 virus, highlights characteristics of viral infection in permissive and non-permissive systems, and focuses on recent findings concerning the pathogenic role of B19V in rheumatologic diseases.
Collapse
Affiliation(s)
- Rosaria Arvia
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (M.A.S.); (K.Z.)
| | - Maria A. Stincarelli
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (M.A.S.); (K.Z.)
| | - Elisabetta Manaresi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (E.M.); (G.G.)
| | - Giorgio Gallinella
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (E.M.); (G.G.)
- S. Orsola-Malpighi Hospital—Microbiology, 40138 Bologna, Italy
| | - Krystyna Zakrzewska
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (M.A.S.); (K.Z.)
| |
Collapse
|
5
|
Jacquot R, Gerfaud-Valentin M, Mekki Y, Billaud G, Jamilloux Y, Sève P. [Parvovirus B19 infections in adults]. Rev Med Interne 2022; 43:713-726. [PMID: 36088203 DOI: 10.1016/j.revmed.2022.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 08/21/2022] [Indexed: 12/11/2022]
Abstract
Acute Parvovirus B19 (PVB19) infection is responsible for erythema infectiosum in children and non-specific polyarthralgias in immunocompetent adults associated with skin lesions and rarer manifestations (hepatic, neurological, cardiac or nephrological). In immunocompromised patients, cytopenias are more frequent and in some cases, viremia persists and is responsible for PVB19 chronic infection. PVB19 is responsible for pure red cell aplasia during chronic hemolytic diseases. Acute PVB19 infection is a differential diagnosis of some autoimmune diseases and has been suspected to be a trigger for some autoimmune diseases because of its ability to promote the emergence of autoimmune markers. Mechanisms of molecular mimicry, induction of apoptosis and activation of enzymes have been demonstrated, explaining in part the production of autoantibodies during infection. However, the demonstration of a causal relationship in the triggering of autoimmune disease remains to be done. This review provides a synthesis of the PVB19 infection clinical data in adults with a particular focus on these links with autoimmunity.
Collapse
Affiliation(s)
- R Jacquot
- Service de médecine interne, hospices civils de Lyon, hôpital de la Croix-Rousse, 103, Grande-Rue de la Croix-Rousse, 69317 Lyon cedex 04, France; Université de Lyon, Lyon, France.
| | - M Gerfaud-Valentin
- Service de médecine interne, hospices civils de Lyon, hôpital de la Croix-Rousse, 103, Grande-Rue de la Croix-Rousse, 69317 Lyon cedex 04, France; Université de Lyon, Lyon, France
| | - Y Mekki
- Université de Lyon, Lyon, France
| | | | - Y Jamilloux
- Service de médecine interne, hospices civils de Lyon, hôpital de la Croix-Rousse, 103, Grande-Rue de la Croix-Rousse, 69317 Lyon cedex 04, France; Université de Lyon, Lyon, France
| | - P Sève
- Service de médecine interne, hospices civils de Lyon, hôpital de la Croix-Rousse, 103, Grande-Rue de la Croix-Rousse, 69317 Lyon cedex 04, France; Université de Lyon, Lyon, France; Université Claude-Bernard Lyon 1, Research on Healthcare Performance (RESHAPE), Inserm U1290, Lyon, France; Laboratoire de virologie, hospices civils de Lyon, centre de biologie et de pathologie, hôpital de la Croix-Rousse, 103, Grande-Rue de la Croix-Rousse, 69317 Lyon cedex 04, France
| |
Collapse
|
6
|
Zhang J, Li P, Zeng H, Huang Y, Hong W. Highly efficient and green synthesis of 2,4-diphenyl substituted thiazoles. SYNTHETIC COMMUN 2020. [DOI: 10.1080/00397911.2020.1718711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Jungan Zhang
- School of Chemistry and Chemical Engineering, North Minzu University, Yinchuan, P. R. China
- Key Laboratory of Chemical Engineering and Technology, State Ethnic Affairs Commission, North Minzu University, Yinchuan, P. R. China
| | - Peipei Li
- School of Pharmacy, Ningxia Medical University, Yinchuan, P. R. China
| | - Hongyun Zeng
- School of Chemistry and Chemical Engineering, North Minzu University, Yinchuan, P. R. China
- Key Laboratory of Chemical Engineering and Technology, State Ethnic Affairs Commission, North Minzu University, Yinchuan, P. R. China
| | - Yu Huang
- School of Pharmacy, Ningxia Medical University, Yinchuan, P. R. China
| | - Wei Hong
- School of Chemistry and Chemical Engineering, North Minzu University, Yinchuan, P. R. China
- Key Laboratory of Chemical Engineering and Technology, State Ethnic Affairs Commission, North Minzu University, Yinchuan, P. R. China
| |
Collapse
|
7
|
Sun F, Chen Y, Wu W, Guo L, Xu W, Chen J, Sun S, Li J, Chen Z, Gu L, Wang X, Li T, Ye S. Varicella zoster virus infections increase the risk of disease flares in patients with SLE: a matched cohort study. Lupus Sci Med 2019; 6:e000339. [PMID: 31413853 PMCID: PMC6667776 DOI: 10.1136/lupus-2019-000339] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/22/2019] [Accepted: 07/17/2019] [Indexed: 11/13/2022]
Abstract
Objective To explore whether varicella zoster virus (VZV) infection could increase the risk of disease flares in patients with SLE. Methods Patients who had VZV reactivations between January 2013 and April 2018 were included from the SLE database (n=1901) of Shanghai Ren Ji Hospital, South Campus. Matched patients with SLE were selected as background controls with a 3:1 ratio. Patients with SLE with symptomatic bacterial infections of the lower urinary tract (UTI) were identified as infection controls. Baseline period and index period were defined as 3 months before and after infection event, respectively. Control period was the following 3 months after the index period. Flare was defined by SELENA SLEDAI Flare Index. Kaplan-Meier analysis, Cox regression model and propensity score weighting were applied. Results Patients with VZV infections (n=47), UTI controls (n=28) and matched SLE background controls (n=141) were included. 16 flares (34%) in the VZV group within the index period were observed, as opposed to only 7.1% in UTI controls and 9.9% in background controls. Kaplan-Meier curve revealed that patients with a VZV infection had a much lower flare-free survival within the index period compared with the controls (p=0.0003). Furthermore, after adjusting for relevant confounders including baseline disease activity and intensity of immunosuppressive therapy, Cox regression analysis and propensity score weighting confirmed that VZV infection within 3 months was an independent risk factor for SLE flares (HR 3.70 and HR 4.16, respectively). Conclusions In patients with SLE, recent VZV infection within 3 months was associated with increased risk of disease flares.
Collapse
Affiliation(s)
- Fangfang Sun
- Department of Rheumatology, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.,Department of Emergency, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yi Chen
- Department of Emergency, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wanlong Wu
- Department of Rheumatology, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Li Guo
- Department of Rheumatology, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wenwen Xu
- Department of Rheumatology, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jie Chen
- Department of Rheumatology, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shuhui Sun
- Department of Rheumatology, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jiajie Li
- Department of Rheumatology, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zhiwei Chen
- Department of Rheumatology, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Liyang Gu
- Department of Rheumatology, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaodong Wang
- Department of Rheumatology, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ting Li
- Department of Rheumatology, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shuang Ye
- Department of Rheumatology, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
8
|
Hod T, Zandman-Goddard G, Langevitz P, Rudnic H, Grossman Z, Rotman-Pikielny P, Levy Y. Does parvovirus infection have a role in systemic lupus erythematosus? Immunol Res 2017; 65:447-453. [DOI: 10.1007/s12026-017-8896-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
9
|
Kerr JR. The role of parvovirus B19 in the pathogenesis of autoimmunity and autoimmune disease. J Clin Pathol 2015; 69:279-91. [PMID: 26644521 DOI: 10.1136/jclinpath-2015-203455] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/06/2015] [Indexed: 11/03/2022]
Abstract
Human parvovirus B19 is a single-stranded DNA virus which preferentially targets the erythroblasts in the bone marrow. B19 infection commonly causes erythema infectiosum, arthralgia, fetal death, transient aplastic crisis in patients with shortened red cell survival, and persistent infection in people who are immunocompromised. Less common clinical manifestations include atypical skin rashes, neurological syndromes, cardiac syndromes, and various cytopenias. B19 infection has also been associated with development of a variety of different autoimmune diseases, including rheumatological, neurological, neuromuscular, cardiovascular, haematological, nephrological and metabolic. Production of a variety of autoantibodies has been demonstrated to occur during B19 infection and these have been shown to be key to the pathogenesis of the particular disease process in a significant number of cases, for example, production of rheumatoid factor in cases of B19-associated rheumatoid arthritis and production of anti-glutamic acid decarboxylase (GAD) in patients with B19-associated type 1 diabetes mellitus. B19 infection has also been associated with the development of multiple autoimmune diseases in 12 individuals. Documented mechanisms in B19-associated autoimmunity include molecular mimicry (IgG antibody to B19 proteins has been shown to cross react with a variety of recognised human autoantigens, including collagen II, keratin, angiotensin II type 1 receptor, myelin basic protein, cardiolipin, and platelet membrane glycoprotein IIb/IIIa), B19-induced apoptosis with presentation of self-antigens to T lymphocytes, and the phospholipase activity of the B19 unique VP1 protein.
Collapse
|
10
|
Page C, François C, Goëb V, Duverlie G. Human parvovirus B19 and autoimmune diseases. Review of the literature and pathophysiological hypotheses. J Clin Virol 2015; 72:69-74. [DOI: 10.1016/j.jcv.2015.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/04/2015] [Accepted: 09/22/2015] [Indexed: 10/23/2022]
|
11
|
Ribeiro FM, Gomez VE, Albuquerque EMN, Klumb EM, Shoenfeld Y. Lupus and leprosy: beyond the coincidence. Immunol Res 2015; 61:160-3. [PMID: 25412733 DOI: 10.1007/s12026-014-8596-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Systemic lupus erythematous (SLE) is an autoimmune disease that presents an increased susceptibility to infections which may trigger reactivation. Disease flares have been mostly associated with parvovirus B19, cytomegalovirus, EBV and Mycobacterium tuberculosis infections, but it is probable that many other agents may also induce innate and adaptive immune system stimulation including the production of autoantibodies as ANA, anti nDNA and anti-ß2-GPI mainly in lepromatous leprosy. Mycobacterium leprae not only may determine symptoms that mimic lupus flares, including autoantibodies production, but could also act as a trigger for lupus reactivation; however, its association is still not fully explored. As demonstrated for tuberculosis, it is quite possible that molecular mimicry may also be involved in the interface of these two diseases. Some studies reported shared epitopes among idiotypes derived from 8E7 and TH9 lepromatous antibodies and those obtained from SLE patients, and it could partially explain the triggering phenomenon of SLE caused by M. leprae. We report and discuss three Brazilian patients whose disease was inactive and presented disease flares concurrently with the diagnosis of leprosy.
Collapse
Affiliation(s)
- F M Ribeiro
- Disciplina de Reumatologia, Universidade do Estado do Rio de Janeiro, Rua Dois de Dezembro, 131/702 - Flamengo, Rio de Janeiro, RJ, 22220-040, Brazil,
| | | | | | | | | |
Collapse
|
12
|
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by multisystem organ involvement, heterogeneity of clinical features, and variety in degree of severity. The differential diagnosis is a crucial aspect in SLE as many other autoimmune diseases portray clinical similarities and autoantibody positivity. Lupus mimickers refer to a group of conditions that exhibit both clinical features and laboratory characteristics, including autoantibody profiles that resemble those present in patients with SLE, and prompt a diagnostic challenge in everyday clinical practice. Thus, lupus mimickers may present as a lupus-like condition (i.e., 2 or 3 criteria) or as one meeting the classification criteria for SLE. Herein we review and classify the current literature on lupus mimickers based on diverse etiologies which include infections, malign and benign neoplasms, medications, and vaccine-related reactions.
Collapse
Affiliation(s)
- Omar-Javier Calixto
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia; Mederi, Hospital Universitario Mayor, Bogotá, Colombia
| | - Juan-Sebastian Franco
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia; Mederi, Hospital Universitario Mayor, Bogotá, Colombia
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia; Mederi, Hospital Universitario Mayor, Bogotá, Colombia.
| |
Collapse
|
13
|
Ahmed SS, Schur PH, MacDonald NE, Steinman L. Narcolepsy, 2009 A(H1N1) pandemic influenza, and pandemic influenza vaccinations: What is known and unknown about the neurological disorder, the role for autoimmunity, and vaccine adjuvants. J Autoimmun 2014; 50:1-11. [DOI: 10.1016/j.jaut.2014.01.033] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 01/13/2014] [Accepted: 01/22/2014] [Indexed: 11/17/2022]
|
14
|
Abstract
We recently introduced the concept of the infectome as a means of studying all infectious factors which contribute to the development of autoimmune disease. It forms the infectious part of the exposome, which collates all environmental factors contributing to the development of disease and studies the sum total of burden which leads to the loss of adaptive mechanisms in the body. These studies complement genome-wide association studies, which establish the genetic predisposition to disease. The infectome is a component which spans the whole life and may begin at the earliest stages right up to the time when the first symptoms manifest, and may thus contribute to the understanding of the pathogenesis of autoimmunity at the prodromal/asymptomatic stages. We provide practical examples and research tools as to how we can investigate disease-specific infectomes, using laboratory approaches employed from projects studying the “immunome” and “microbiome”. It is envisioned that an understanding of the infectome and the environmental factors that affect it will allow for earlier patient-specific intervention by clinicians, through the possible treatment of infectious agents as well as other compounding factors, and hence slowing or preventing disease development.
Collapse
|
15
|
Thammasri K, Rauhamäki S, Wang L, Filippou A, Kivovich V, Marjomäki V, Naides SJ, Gilbert L. Human parvovirus B19 induced apoptotic bodies contain altered self-antigens that are phagocytosed by antigen presenting cells. PLoS One 2013; 8:e67179. [PMID: 23776709 PMCID: PMC3680405 DOI: 10.1371/journal.pone.0067179] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 05/15/2013] [Indexed: 12/03/2022] Open
Abstract
Human parvovirus B19 (B19V) from the erythrovirus genus is known to be a pathogenic virus in humans. Prevalence of B19V infection has been reported worldwide in all seasons, with a high incidence in the spring. B19V is responsible for erythema infectiosum (fifth disease) commonly seen in children. Its other clinical presentations include arthralgia, arthritis, transient aplastic crisis, chronic anemia, congenital anemia, and hydrops fetalis. In addition, B19V infection has been reported to trigger autoimmune diseases such as systemic lupus erythematosus and rheumatoid arthritis. However, the mechanisms of B19V participation in autoimmunity are not fully understood. B19V induced chronic disease and persistent infection suggests B19V can serve as a model for viral host interactions and the role of viruses in the pathogenesis of autoimmune diseases. Here we investigate the involvement of B19V in the breakdown of immune tolerance. Previously, we demonstrated that the non-structural protein 1 (NS 1) of B19V induces apoptosis in non-permissive cells lines and that this protein can cleave host DNA as well as form NS1-DNA adducts. Here we provide evidence that through programmed cell death, apoptotic bodies (ApoBods) are generated by B19V NS1 expression in a non-permissive cell line. Characterization of purified ApoBods identified potential self-antigens within them. In particular, signature self-antigens such as Smith, ApoH, DNA, histone H4 and phosphatidylserine associated with autoimmunity were present in these ApoBods. In addition, when purified ApoBods were introduced to differentiated macrophages, recognition, engulfment and uptake occurred. This suggests that B19V can produce a source of self-antigens for immune cell processing. The results support our hypothesis that B19V NS1-DNA adducts, and nucleosomal and lysosomal antigens present in ApoBods created in non-permissive cell lines, are a source of self-antigens.
Collapse
Affiliation(s)
- Kanoktip Thammasri
- Department of Biological and Environmental Sciences and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Sanna Rauhamäki
- Department of Biological and Environmental Sciences and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Liping Wang
- Department of Biological and Environmental Sciences and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Artemis Filippou
- Department of Biological and Environmental Sciences and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Violetta Kivovich
- Pennsylvania State College of Medicine/Milton S. Hershey Medical Center, Hershey, Pennsylvania, United States of America
| | - Varpu Marjomäki
- Department of Biological and Environmental Sciences and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Stanley J. Naides
- Quest Diagnostics Nichols Institute, San Juan Capistrano, California, United States of America
| | - Leona Gilbert
- Department of Biological and Environmental Sciences and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
- * E-mail:
| |
Collapse
|
16
|
Bogdanos DP, Smyk DS, Invernizzi P, Rigopoulou EI, Blank M, Pouria S, Shoenfeld Y. Infectome: a platform to trace infectious triggers of autoimmunity. Autoimmun Rev 2012; 12:726-40. [PMID: 23266520 PMCID: PMC7105216 DOI: 10.1016/j.autrev.2012.12.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 12/12/2012] [Indexed: 02/06/2023]
Abstract
The "exposome" is a term recently used to describe all environmental factors, both exogenous and endogenous, which we are exposed to in a lifetime. It represents an important tool in the study of autoimmunity, complementing classical immunological research tools and cutting-edge genome wide association studies (GWAS). Recently, environmental wide association studies (EWAS) investigated the effect of environment in the development of diseases. Environmental triggers are largely subdivided into infectious and non-infectious agents. In this review, we introduce the concept of the "infectome", which is the part of the exposome referring to the collection of an individual's exposures to infectious agents. The infectome directly relates to geoepidemiological, serological and molecular evidence of the co-occurrence of several infectious agents associated with autoimmune diseases that may provide hints for the triggering factors responsible for the pathogenesis of autoimmunity. We discuss the implications that the investigation of the infectome may have for the understanding of microbial/host interactions in autoimmune diseases with long, pre-clinical phases. It may also contribute to the concept of the human body as a superorganism where the microbiome is part of the whole organism, as can be seen with mitochondria which existed as microbes prior to becoming organelles in eukaryotic cells of multicellular organisms over time. A similar argument can now be made in regard to normal intestinal flora, living in symbiosis within the host. We also provide practical examples as to how we can characterise and measure the totality of a disease-specific infectome, based on the experimental approaches employed from the "immunome" and "microbiome" projects.
Collapse
Affiliation(s)
- Dimitrios P Bogdanos
- Institute of Liver Studies, King's College London School of Medicine at King's College Hospital, Denmark Hill Campus, London, UK.
| | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Despite the very low risk-to-benefit ratio of vaccines, fear of negative side effects has discouraged many people from getting vaccinated, resulting in reemergence of previously controlled diseases such as measles, pertussis, and diphtheria. Part of this fear stems from the lack of public awareness of the many preclinical and clinical safety evaluations that vaccines must undergo before they are available to the general public, as well as from misperceptions of what adjuvants are or why they are used in vaccines. The resultant "black box" leads to a preoccupation with rare side effects (such as autoimmune diseases) that are speculated, but not proven, to be linked to some vaccinations. The focus of this review article is to open this black box and provide a conceptual framework for how vaccine safety is traditionally assessed. We discuss the strengths and shortcomings of tools that can be and are used preclinically (in animal studies), translationally (in biomarker studies with human sera or cells), statistically (for disease epidemiology), and clinically (in the design of human trials) to help ascertain the risk of the infrequent and delayed adverse events that arise in relation to adjuvanted vaccine administration.
Collapse
Affiliation(s)
- S Sohail Ahmed
- Global Clinical Sciences, Vaccines Research, Novartis Vaccines and Diagnostics, 53100 Siena, Italy.
| | | | | | | |
Collapse
|
18
|
Abstract
Since its discovery, human parvovirus B19 (B19V), now termed erythrovirus, has been associated with many clinical situations (neurological and myocardium infections, persistent B19V DNAemia) in addition to the prototype clinical manifestations, i.e., erythema infectiosum and erythroblastopenia crisis. In 2002, the use of new molecular tools led to the characterization of three different genotypes of human B19 erythrovirus. Although the genomic organization is conserved, the geographic distribution of the different genotypes varies worldwide, and the nucleotidic divergences can impact the molecular diagnosis of B19 virus infection. The cell cycle of the virus remains partially unresolved; however, recent studies have shed light on the mechanism of cell entry and the interactions of B19V proteins with apoptosis pathways.
Collapse
|
19
|
Francis L, Perl A. Infection in systemic lupus erythematosus: friend or foe? INTERNATIONAL JOURNAL OF CLINICAL RHEUMATOLOGY 2010; 5:59-74. [PMID: 20209114 PMCID: PMC2830655 DOI: 10.2217/ijr.09.72] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Infectious agents have long been implicated in the pathogenesis of systemic lupus erythematosus. Common viruses, such as the Epstein-Barr virus, transfusion transmitted virus, parvovirus and cytomegalovirus, have an increased prevalence in patients with systemic lupus erythematosus. They may contribute to disease pathogenesis through triggering autoimmunity via structural or functional molecular mimicry, encoding proteins that induce cross-reactive immune responses to self antigens or modulate antigen processing, activation, or apoptosis of B and T cells, macrophages or dendritic cells. Alternatively, some infectious agents, such as malaria, Toxoplasma gondii and Helicobacter pylori, may have a protective effect. Vaccinations may play dual roles by protecting against friend and foe alike.
Collapse
Affiliation(s)
- Lisa Francis
- Division of Rheumatology, Department of Medicine State University of New York, College of Medicine 750 East Adams Street Syracuse, New York 13210, USA
| | - Andras Perl
- Division of Rheumatology, Department of Medicine State University of New York, College of Medicine 750 East Adams Street Syracuse, New York 13210, USA
| |
Collapse
|
20
|
Ramos-Casals M, Cuadrado MJ, Alba P, Sanna G, Brito-Zerón P, Bertolaccini L, Babini A, Moreno A, D'Cruz D, Khamashta MA. Acute viral infections in patients with systemic lupus erythematosus: description of 23 cases and review of the literature. Medicine (Baltimore) 2008; 87:311-318. [PMID: 19011502 DOI: 10.1097/md.0b013e31818ec711] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Few studies have evaluated the impact of viral infections on the daily management of patients with systemic lupus erythematosus (SLE). We analyzed the etiology and clinical features of acute viral infections arising in patients with SLE and their influence on the diagnosis, prognosis, and treatment of SLE. Cases occurring within the last 5 years were selected from the databases of 3 large teaching hospitals. Acute viral infections were confirmed by the identification of specific antiviral IgM antibodies and subsequent seroconversion with detection of specific IgG antibodies. In autopsy studies, macroscopic findings suggestive of viral infection were confirmed by direct identification of the virus or viruses in tissue samples. We performed a MEDLINE search for additional cases reported between January 1985 and March 2008. We included 88 cases (23 from our clinics and 65 from the literature review) of acute viral infections in patients with SLE. Twenty-five patients were diagnosed with new-onset SLE (fulfillment of the 1997 SLE criteria) associated with infection by human parvovirus B19 (n = 15), cytomegalovirus (CMV; n = 6), Epstein-Barr virus (EBV; n = 3), and hepatitis A virus (n = 1). The remaining 63 cases of acute viral infections arose in patients already diagnosed with SLE: in 18 patients, symptoms related to infection mimicked a lupus flare, 36 patients, including 1 patient from the former group who presented with both conditions, presented organ-specific viral infections (mainly pneumonitis, colitis, retinitis, and hepatitis), and 10 patients presented a severe, multiorgan process similar to that described in catastrophic antiphospholipid syndrome-the final diagnosis was hemophagocytic syndrome in 5 cases and disseminated viral infection in 5. Twelve patients died due to infection caused by CMV (n = 5), herpes simplex virus (n = 4), EBV (n = 2), and varicella zoster virus (n = 1). Autopsies were performed in 9 patients and disclosed disseminated herpetic infection in 6 patients (caused by herpes simplex in 4 cases, varicella in 1, and CMV in 1) and hemophagocytic syndrome in 3. A higher frequency of renal failure (54% vs. 19%, p = 0.024), antiphospholipid syndrome (33% vs. 6%, p = 0.023), treatment with cyclophosphamide (82% vs. 37%, p = 0.008), and multisystemic involvement at presentation (58% vs. 8%, p < 0.001); and a lower frequency of antiviral therapy (18% vs. 76%, p < 0.001) were found in patients who died, compared with survivors. The most common viral infections in patients with SLE are parvovirus B19 (predominantly mimicking SLE presentation) and CMV (predominantly presenting in severely immunosuppressed patients). CMV infection may mimic a lupus flare or present with specific organ involvement such as gastrointestinal bleeding or pulmonary infiltrates. Other herpesviruses are common in immunosuppressed SLE patients and may produce a wide range of manifestations. Physicians should examine the pharynx, eyes, skin, and genitalia and should conduct serologic and molecular studies to improve early detection of viral infection in patients with SLE.
Collapse
Affiliation(s)
- Manuel Ramos-Casals
- From Laboratory of Autoimmune Diseases "Josep Font," Department of Autoimmune Diseases (MR-C, PB-Z) and Department of Infectious Diseases (AM), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain; Department of Rheumatology (PA, AB), Hospital Córdoba, Córdoba, Argentina; Department of Rheumatology (GS), Homerton University NHS Foundation Trust, London, United Kingdom; and Lupus Research Unit (MJC, LB, DD'C, MAK), The Rayne Institute, King's College London School of Medicine at Guy's, King's and St Thomas' Hospitals, St Thomas' Hospital, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Pordeus V, Szyper-Kravitz M, Levy RA, Vaz NM, Shoenfeld Y. Infections and autoimmunity: a panorama. Clin Rev Allergy Immunol 2008; 34:283-99. [PMID: 18231878 PMCID: PMC7090595 DOI: 10.1007/s12016-007-8048-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
For more than 2,000 years, it was thought that malignant spirits caused diseases. By the end of nineteenth century, these beliefs were displaced by more modern concepts of disease, namely, the formulation of the “germ theory,” which asserted that bacteria or other microorganisms caused disease. With the emergence of chronic degenerative and of autoimmune diseases in the last century, the causative role of microorganisms has been intensely debated; however, no clear explanatory models have been achieved. In this review, we examine the current available literature regarding the relationships between infections and 16 autoimmune diseases. We critically analyzed clinical, serological, and molecular associations, and reviewed experimental models of induction of and, alternatively, protection from autoimmune diseases by infection. After reviewing several studies and reports, a clinical and experimental pattern emerges: Chronic and multiple infections with viruses, such as Epstein–Barr virus and cytomegalovirus, and bacteria, such as H. pylori, may, in susceptible individuals, play a role in the evolvement of autoimmune diseases. As the vast majority of infections pertain to our resident microbiota and endogenous retroviruses and healthy carriage of infections is the rule, we propose to focus on understanding the mechanisms of this healthy carrier state and what changes its configurations to infectious syndromes, to the restoration of health, or to the sustaining of illness into a chronic state and/or autoimmune disease. It seems that in the development of this healthy carriage state, the infection or colonization in early stages of ontogenesis with key microorganisms, also called ‘old friends’ (lactobacilli, bifidobacteria among others), are important for the healthy living and for the protection from infectious and autoimmune syndromes.
Collapse
Affiliation(s)
- V Pordeus
- Clinical Research, Pro Cardiaco Hospital Research Center-PROCEP, Rio de Janeiro, Brazil
| | | | | | | | | |
Collapse
|
22
|
|
23
|
Drechsler MD, Obermeier I, Döring Y, Lackner KJ, Modrow S, von Landenberg P. Generation of multifunctional murine monoclonal antibodies specifically directed to the VP1unique region protein of human parvovirus B19. Immunobiology 2008; 213:511-7. [PMID: 18514753 DOI: 10.1016/j.imbio.2007.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Revised: 12/05/2007] [Accepted: 12/13/2007] [Indexed: 11/17/2022]
Abstract
Little is known about the VP1unique region (VP1u), a part of one major capsid protein of human parvovirus B19 (B19), concerning its involvement in viral replication and infection cycle. Showing a phospholipase A2 (PLA2)-like activity, which is discussed to be necessary for viral release from host cell, its precise function remains unclear. The purpose of this study was to generate multifunctional monoclonal antibodies (mabs) for different applications that may be useful in investigating VP1u's relevance. To establish antiVP1u antibodies, spleen cells from Balb/c mice immunized with purified recombinant viral protein were used for generating antibody-producing hybridoma cell lines. Usability of the antibodies was tested in enzyme-linked immunosorbent assay (ELISA), Western-blot analysis, immunofluorescence and an inhibition assay of enzymatic activity of PLA2. Three hybridoma cell lines secreting mab's specifically directed against the VP1u protein of B19 could be generated and functioned in every screening method used in this study. These antibodies are helpful tools for investigations in B19 research and diagnosis. Furthermore, the antibodies could help in gaining a deeper understanding of VP1u's role in viral replication and infection especially in the importance of its constitutive PLA2-like activity.
Collapse
Affiliation(s)
- Maik D Drechsler
- Institute of Clinical Chemistry and Laboratory Medicine, Johannes Gutenberg-Universität Mainz Klinikum, Langenbeckstrasse 1, 55131 Mainz, Germany
| | | | | | | | | | | |
Collapse
|
24
|
Franssila R, Hedman K. Infection and musculoskeletal conditions: Viral causes of arthritis. Best Pract Res Clin Rheumatol 2007; 20:1139-57. [PMID: 17127201 DOI: 10.1016/j.berh.2006.08.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Several viruses cause postinfectious arthritis. The disease is a typical manifestation of arthritogenic alphaviruses, rubella virus and human parvovirus B19. In addition, arthritis is not uncommon after infection by HIV, cytomegalovirus, hepatitis B virus, hepatitis C virus, or Epstein-Barr virus (EBV). Also prolonged arthritis may result from viral infections, particularly with alphaviruses and human parvovirus B19. Viruses such as EBV and B19 may have significant roles in initiating chronic arthropathies, which in some cases may be indistinguishable from rheumatoid arthritis.
Collapse
Affiliation(s)
- Rauli Franssila
- Department of Virology, Haartman Institute, University of Helsinki and HUCH Laboratory Division, Haartmaninkatu 3, FI-00290 Helsinki, Finland.
| | | |
Collapse
|
25
|
Baskan EB, Yilmaz E, Saricaoglu H, Alkan G, Ercan I, Mistik R, Adim SB, Goral G, Dilek K, Tunali S. Detection of parvovirus B19 DNA in the lesional skin of patients with Behçet's disease. Clin Exp Dermatol 2007; 32:186-90. [PMID: 17250756 DOI: 10.1111/j.1365-2230.2006.02330.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND There is disagreement in the current evidence for viral aetiologies in the pathogenesis of Behçet's disease (BD). OBJECTIVES To investigate the presence of B19 DNA in skin lesions of patients with BD, compare with the skin of healthy controls and evaluate its role in the pathogenesis. METHODS In total, 40 patients diagnosed with BD according to the criteria proposed by the International Study Group for Behçet's Disease and routinely followed up at our centre were enrolled into the study. All the patients selected were in the active phase of disease. Skin and blood samples of patients with BD and of the healthy volunteers were examined for B19 serology, histopathology and genome expression. RESULTS The quantity of B19 DNA in nonulcerative BD lesions of was significantly different from ulcerative lesions in the study group and from the skin of the healthy controls (P < 0.01). For the nonulcerative lesions, real-time PCR analysis for B19 DNA was found to be 64% sensitive (95% CI 42.5-82.0) and 85% specific (95% CI 62.1-96.6) with a cut-off value of > 154 IU/mL (P < 0.001). CONCLUSIONS To the best of our knowledge, this is the first study that provides evidence for a possible causal link between BD and parvovirus B19, and our data suggest the presence of the virus, particularly in intact, nonulcerative skin lesions of BD. Limitations to this study include the limited number of participants, and the fact that the exact source of B19 DNA was undetected.
Collapse
Affiliation(s)
- E B Baskan
- Department of Dermatology, Uludag University Medical Faculty, 16059 Gorukle, Bursa, Turkey.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Modrow S. Parvovirus B19: the causative agent of dilated cardiomyopathy or a harmless passenger of the human myocard? ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2006:63-82. [PMID: 16329658 DOI: 10.1007/3-540-30822-9_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Parvovirus B19 infections may cause a widespread benign and self-limiting disease in children and adults known as erythema infectiosum (fifth disease). Several further manifestations are associated with B19 infections, such as arthralgias, arthritis, leucopenia and thrombocytopenia, anaemia and vasculitis and spontaneous abortion and hydrops fetalis in pregnant women. Persistent infections with continuous virus production may occur in immunocompetent as well as in immunosuppressed individuals. Parvovirus B19 infections have been frequently implicated as a cause or trigger of various forms of autoimmune diseases affecting joints, connective tissue and large and small vessels. Autoimmune neutropenia, thrombocytopenia and haemolytic anaemia are known as sequelae of B19 infections. The molecular basis of the autoimmune phenomena is unclear. Many patients with these long-lasting symptoms are not capable of eliminating the virus or controlling its propagation. Furthermore, latent viral genomes have been detected in cells of various organs and tissues by PCR. At present, it is not clear if these cells produce viral proteins and/or infectious B19 particles, if the virus genome can be reactivated to productive replication and if the presence of viral DNA indicates a causative role of parvovirus B19 with distinct diseases.
Collapse
Affiliation(s)
- S Modrow
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Regensburg, Germany.
| |
Collapse
|
27
|
Lindner J, Barabas S, Saar K, Altmann D, Pfister A, Fleck M, Deml L, Modrow S. CD4+ T-Cell Responses Against the VP1-Unique Region in Individuals with Recent and Persistent Parvovirus B19 Infection. ACTA ACUST UNITED AC 2005; 52:356-61. [PMID: 16316401 DOI: 10.1111/j.1439-0450.2005.00860.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To date cellular immune responses against parvovirus B19 (B19) have not been studied extensively. The aim of this study was to examine the T-cell response against the VP1-unique region as the immunodominant part of the viral structural protein VP1 in individuals with different courses of B19 infection. Therefore, a group of 13 parvovirus-positive probands was separated into subgroups characterized for recent or acute, past or persistent infection by means of the presence of specific immunoglobulin (Ig)M and IgG isotypes and of viral DNA in blood and tissue. Transiently transfected B-cells expressing VP1-unique region were used in ELISpot assays to investigate T-cell responses directed against the VP1-unique region in peripheral blood mononuclear cells (PBMC) of individual donors. Significant numbers of interferon-gamma (IFN-gamma) secreting lymphocytes were detectable in PBMC of all individuals with recent, acute or persistent B19 infection, but not in PBMC of donors with past B19 infection and seronegative individuals. A more detailed analysis of IFN-gamma producing cells by intracellular cytokine staining by flow cytometry revealed, that CD4(+) T cells but not CD8(+) cytotoxic lymphocytes (CTL) were the major subpopulation of IFN-gamma producing cells. These data strongly suggest the need of virus protein production for the maintenance of VP1-unique region-specific CD4(+) T-helper cell responses in B19-infected individuals.
Collapse
Affiliation(s)
- J Lindner
- Institute of Medical Microbiology and Hygiene, University of Regensburg, 93053 Regensburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Sève P, Ferry T, Koenig M, Cathebras P, Rousset H, Broussolle C. Lupus-like presentation of parvovirus B19 infection. Semin Arthritis Rheum 2005; 34:642-8. [PMID: 15692957 DOI: 10.1016/j.semarthrit.2004.07.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVES To describe 2 cases of parvovirus B19 (B19) infection mimicking systemic lupus erythematosus (SLE) and to identify all cases of SLE imitated by and/or associated with B19 in the medical literature. METHODS A computer-assisted (PubMed) search of the medical literature from 1975 to 2003 was performed using the following key words: parvovirus, B19, SLE, lupus, antibodies, auto-immunity. RESULTS Thirty-eight patients were identified: 35 women, 3 men; mean age = 28.8 years. Clinical manifestations were as follows: fever (24 patients); articular involvement (36 patients); cutaneous lesions (28 patients); lymphadenopathy (9 patients); hepato- and/or splenomegaly (6 patients); serositis (6 patients); renal involvement (4 patients); cerebral impairment (10 patients). Cytopenia was observed in 23 cases. Antinuclear antibodies were detected in 34 patients, anti-double-stranded DNA antibodies in 20 patients, anti-Sm antibodies in 4 patients, antinuclear ribonucleoprotein antibodies in 5 patients, anti-Ro-SSA antibodies in 4 patients, anti-La-SSB antibodies in 4 patients, and anticardiolipin and/or anti-beta2-glycoprotein I antibodies in 8 patients. Hypocomplementemia was found in 15 of 26 patients. In 19 cases, the B19 infection had a self-limiting course. In 6 cases, B19 infection occurred in a context of previously established SLE, simulating SLE exacerbation. In 6 observations, symptoms persisted several months after the viral infection. In 7 cases, the exact relationship between SLE and B19 could not be determined. CONCLUSIONS B19 infection may present a clinical and serological tableau making it difficult to distinguish between a viral infection and the first episode of SLE. Although B19 may modulate the clinical and biological features of rheumatic disease, studies in large series do not support a causative role for B19 in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Pascal Sève
- Department of Internal Medicine, Hôtel-Dieu, Hospices Civils de Lyon, Université Claude Bernard Lyon I, France.
| | | | | | | | | | | |
Collapse
|
29
|
Sève P, Ferry T, Charhon A, Calvet A, Broussolle C. Manifestations systémiques des infections à Parvovirus B19. Rev Med Interne 2004; 25:740-51. [PMID: 15471600 DOI: 10.1016/j.revmed.2004.02.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2003] [Accepted: 02/26/2004] [Indexed: 11/30/2022]
Abstract
PURPOSE Parvovirus B19 (B19) causes many clinical disorders, of which the most common are erythema infectiosum, aplastic crisis complicating chronic hemolytic anemia, and hydrops fetalis. In young adults, the skin eruption caused by B19 is accompanied by polyarthritis and polyarthralgia in 60% of the cases. Rheumatoid factors and other antibodies including antinuclear antibodies, anti-ADN, and antiphospholipids can be produced in the wake of B19 infection. CURRENT KNOWLEDGE AND KEY POINTS These features may simulate systemic diseases as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) (lupus-like eruption over the cheeks, cytopenia, etc.) or vasculitis (purpura, renal involvement). In addition, there have been a few reports of SLE, vasculitis and other connective tissue diseases developing shortly after a B19 infection associated with virus clearance suggesting that B19 can act as a trigger of systemic disease. However, studies in large series indicate that in fact B19 is probably an extremely rare cause of RA, SLE or vasculitis. FUTURE PROSPECTS AND PROJECTS In fundamental studies B19 interacts with inflammatory cells by regulation of cytokines. More recently, two studies suggest that viral infection due to B19 may affect the course of SLE, leading to specific biological subsets. These preliminary findings require confirmation to elucidate the significance of the presence of B19 in systemic disease.
Collapse
MESH Headings
- Adolescent
- Adrenal Cortex Hormones/therapeutic use
- Adult
- Aged
- Antibodies, Viral/analysis
- Arthritis, Infectious/diagnosis
- Arthritis, Infectious/etiology
- Arthritis, Infectious/virology
- Arthritis, Rheumatoid/diagnosis
- Arthritis, Rheumatoid/etiology
- Arthritis, Rheumatoid/virology
- Autoantibodies/analysis
- Autoimmune Diseases/diagnosis
- Autoimmune Diseases/immunology
- Autoimmune Diseases/virology
- Azathioprine/therapeutic use
- Child
- Child, Preschool
- Cyclophosphamide/therapeutic use
- Diagnosis, Differential
- Female
- Humans
- Immunosuppressive Agents/therapeutic use
- Lupus Erythematosus, Systemic/diagnosis
- Lupus Erythematosus, Systemic/etiology
- Lupus Erythematosus, Systemic/virology
- Male
- Middle Aged
- Parvoviridae Infections/complications
- Parvoviridae Infections/diagnosis
- Parvoviridae Infections/drug therapy
- Parvoviridae Infections/immunology
- Parvoviridae Infections/virology
- Parvovirus B19, Human/isolation & purification
- Parvovirus B19, Human/pathogenicity
- Pregnancy
- Pregnancy Complications, Infectious/diagnosis
- Skin Diseases, Viral/diagnosis
- Skin Diseases, Viral/etiology
- Treatment Outcome
- Vasculitis/diagnosis
- Vasculitis/etiology
- Vasculitis/virology
Collapse
Affiliation(s)
- P Sève
- Service de médecine interne, Hôtel-Dieu, 1 place de l'Hôpital, 69288 Lyon cedex 02, France.
| | | | | | | | | |
Collapse
|
30
|
Gallinella G, Venturoli S, Manaresi E, Musiani M, Zerbini M. B19 virus genome diversity: epidemiological and clinical correlations. J Clin Virol 2003; 28:1-13. [PMID: 12927746 DOI: 10.1016/s1386-6532(03)00120-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Genetic analysis of parvovirus B19 has been carried out mainly to establish a framework to track molecular epidemiology of the virus and to correlate sequence variability with different pathological and clinical manifestations of the virus. A good amount of information regarding B19 virus sequence variability is available, and presently there are about 400 sequence records deposited in the nucleotide database of NCBI. A few are almost complete genomic sequences, and these allow the construction of a global alignment framework. Many others are partial genomic sequences, limited to selected regions, and these allow comparison of a higher number of isolates from well-defined epidemiological settings and/or pathological conditions. Most studies showed that the genetic variability of B19 virus is low, that molecular epidemiology is possible only on a limited geographical and temporal setting, and that no clear correlations are present between genome sequence and distinctive pathological and clinical manifestations. More recently, several viral isolates have been identified that show remarkable sequence diversity with respect to reference sequences. The identification of variant isolates added to the knowledge of genetic diversity in this virus group and allowed the identification of three divergent genetic clusters, about 10% divergent from each other and still quite distinct from other parvoviruses, that can be thought of as different genotypes within the human erythrovirus group and that show clearly resolved phylogenetic relationship. These variant isolates pose interesting questions regarding the real extent of genetic variability in the human erythroviruses, the relevance of these viruses in terms of epidemiology and their possible implication in the pathogenesis of erythrovirus-related diseases.
Collapse
Affiliation(s)
- Giorgio Gallinella
- Department of Clinical and Experimental Medicine, Division of Microbiology, University of Bologna, Via Massarenti, 9, I-40138 Bologna, Italy
| | | | | | | | | |
Collapse
|
31
|
Von Landenberg P, Lehmann HW, Knöll A, Dorsch S, Modrow S. Antiphospholipid antibodies in pediatric and adult patients with rheumatic disease are associated with parvovirus B19 infection. ARTHRITIS AND RHEUMATISM 2003; 48:1939-47. [PMID: 12847688 DOI: 10.1002/art.11038] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To show a possible association between parvovirus B19 infection and the presence of antiphospholipid antibodies (aPL) in patients with rheumatic diseases. METHODS Serum samples obtained from 88 children with various forms of juvenile rheumatic disease and from 40 adults with systemic lupus erythematosus, the antiphospholipid syndrome, or other rheumatic disease, who had previously been tested and shown to be positive for IgG aPL, were analyzed for the presence of B19 DNA, for antibodies against the B19 viral proteins VP1, VP2, and NS1, and for IgG aPL (anticardiolipin, anti-beta(2)-glycoprotein I, and antiphosphatidylserine). As controls, serum samples obtained from 135 children with noninflammatory bone diseases or growth retardation were also analyzed. RESULTS Twenty-four (27%) of the 88 children with rheumatic diseases had detectable amounts of IgG aPL. Fourteen (58%) of these 24 IgG aPL-positive patients showed IgG against VP1/VP2 and viral genomes, indicating the presence of acute (2 patients) or persistent (12 patients) infection. Past parvovirus B19 infection was identified in 7 (29%) of 24 IgG aPL-positive children, as indicated by VP1/VP2-specific IgG in the absence of viral DNA. Three (12%) of 24 IgG aPL-positive children had not been infected with B19. Sixty-nine (51%) of 135 control children displayed VP1/VP2-specific IgG. Three (2%) of these 135 children were IgG aPL positive (2 children had past parvovirus B19 infection, and 1 was negative for parvovirus B19). Analysis of the parvovirus B19 status of 40 adult IgG aPL-positive patients showed that 33 (83%) were anti-IgG VP1/VP2-positive, and viral DNA was detected in 11 patients (28%). Ten of these 11 viremic patients were in the subgroup of 28 IgG aPL-positive SLE patients. CONCLUSION Antiphospholipid antibodies are preferentially found in serum of children with juvenile idiopathic arthritis who have been previously infected with parvovirus B19 and have established, persistent infection. Adult patients with IgG aPL positivity have a high incidence of persistent parvovirus B19 infection. We conclude that parvovirus B19 might be directly involved in the elicitation of autoimmune reactions partly mediated by aPL.
Collapse
Affiliation(s)
- Philipp Von Landenberg
- Klinik and Poliklinik für Innere Medizin I, Universitätsklinikum Regensburg, Regensburg, Germany
| | | | | | | | | |
Collapse
|
32
|
Abstract
Human parvovirus B19 infections may cause a widespread benign and self-limiting disease in children and adults, known as erythema infectiosum or fifth disease. A variety of further manifestations are associated with the infection such as arthralgias, arthritis, leukopenia and thrombocytopenia, anemia and vasculitis, spontaneous abortion and hydrops fetalis in pregnant women. Both in children and adults parvovirus B19 infections have been frequently implicated as a cause or trigger of various forms of autoimmune diseases affecting joints, connective tissue and large and small vessels. In addition, autoimmune neutropenia, thrombocytopenia and hemolytic anemia are known as sequelae of B19 infection. The molecular basis of the autoimmune phenomena and resultant pathogenesis is unclear. The involvement of molecular mimicry between cellular and viral proteins, the induction of enhanced cytokine production via the viral transactivator protein NS1 and the phospholipase A2-like activity of the capsid protein VP1 may contribute to the induction of autoimmune reactions. All the known data and the potential mechanisms involved in the pathogenesis will be discussed in this review.
Collapse
Affiliation(s)
- Hartwig W Lehmann
- Klinik für Kinder- und Jugendmedizin, Sächsisches Krankenhaus Hubertusburg, 04779 Wermsdorf, Germany
| | | | | |
Collapse
|
33
|
Abstract
Parvovirus B19 (B19) causes many clinical disorders, of which the most common are erythema infectiosum, aplastic crisis complicating chronic hemolytic anemia, and hydrops fetalis. In young adults, the skin eruption caused by B19 is accompanied with polyarthritis and polyarthralgia in 60% of the cases. The joint abnormalities predominate in the hands and feet and usually resolve within a week (range 2-21 d). Serological tests show IgM antibodies against B19, confirming the diagnosis of recent infection. Protracted polyarthritis occurs in some patients and seems associated with the DR4 histocompatibility alleles. Rheumatoid factors can be produced transiently in these patients. Other autoantibodies produced in the wake of B19 infection include anti-nuclear antibodies, anti-DNA, anti-SSA/SSB, and anti-phospholipids. Acute B19 infection can simulate early rheumatoid arthritis (RA) or systemic lupus erythematosus (SLE) (lupus-like eruption over the cheeks, cytopenia, etc.). In addition, there have been a few reports of erosive RA or SLE developing shortly after a B19 infection, with positive PCR tests for B19 DNA in synovial tissue or blood cells. Studies in large series indicate that B19 is probably an extremely rare cause of RA or SLE. Vasculitides affecting the small vessels (Henoch-Schonlein purpura, Wegener's granulomatosis), medium-sized vessels (periarteritis nodosa), and large vessels (giant cell arteritis) can occur after B19 infection. Here again, the number of clinical cases is small.
Collapse
Affiliation(s)
- Olivier Meyer
- Rheumatology department, Hôpital Bichat, 46, rue Henri-Huchard, 75018 Paris, France.
| |
Collapse
|
34
|
|
35
|
Servant A, Laperche S, Lallemand F, Marinho V, De Saint Maur G, Meritet JF, Garbarg-Chenon A. Genetic diversity within human erythroviruses: identification of three genotypes. J Virol 2002; 76:9124-34. [PMID: 12186896 PMCID: PMC136440 DOI: 10.1128/jvi.76.18.9124-9134.2002] [Citation(s) in RCA: 211] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
B19 virus is a human virus belonging to the genus Erythrovirus: The genetic diversity among B19 virus isolates has been reported to be very low, with less than 2% nucleotide divergence in the whole genome sequence. We have previously reported the isolation of a human erythrovirus isolate, termed V9, whose sequence was markedly distinct (>11% nucleotide divergence) from that of B19 virus. To date, the V9 isolate remains the unique representative of a new variant in the genus Erythrovirus, and its taxonomic position is unclear. We report here the isolation of 11 V9-related viruses. A prospective study conducted in France between 1999 and 2001 indicates that V9-related viruses actually circulate at a significant frequency (11.4%) along with B19 viruses. Analysis of the nearly full-length genome sequence of one V9-related isolate (D91.1) indicates that the D91.1 sequence clusters together with but is notably distant from the V9 sequence (5.3% divergence) and is distantly related to B19 virus sequences (13.8 to 14.2% divergence). Additional phylogenetic analysis of partial sequences from the V9-related isolates combined with erythrovirus sequences available in GenBank indicates that the erythrovirus group is more diverse than thought previously and can be divided into three well-individualized genotypes, with B19 viruses corresponding to genotype 1 and V9-related viruses being distributed into genotypes 2 and 3.
Collapse
Affiliation(s)
- Annabelle Servant
- Laboratoire de Virologie, Hôpital Armand Trousseau, EA2391, UFR Saint-Antoine, Paris, France
| | | | | | | | | | | | | |
Collapse
|
36
|
Mitchell LA. Parvovirus B19 nonstructural (NS1) protein as a transactivator of interleukin-6 synthesis: common pathway in inflammatory sequelae of human parvovirus infections? J Med Virol 2002; 67:267-74. [PMID: 11992589 DOI: 10.1002/jmv.2217] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
This review focuses on the role that human parvovirus B19 nonstructural (NS1) protein as a transactivator of the proinflammatory cytokine, interleukin-6 (IL-6), might play in triggering the multiparametric inflammatory outcomes of B19 infection. Parvovirus B19 is a ubiquitous virus, and it is often expressed during conditions of immunodepression including that induced by long-term chemotherapy, viral infection (HIV, HTLV-1), or genetic immunodeficiency disorders. Through NS1 expression, B19 may contribute to the immune dysregulation associated with these disorders, or serve as a cofactor in enhancing retroviral replication. Hence, NS1 transactivation of proinflammatory cytokine promoters such as IL-6 may be pivotal in triggering the various inflammatory and autoimmune disorders that have been linked to parvovirus B19 infections.
Collapse
|
37
|
Narváez Garcia FJ, Domingo-Domènech E, Castro-Bohorquez FJ, Biosca M, Garcia-Quintana A, Perez-Vega C, Vilaseca-Momplet J. Lupus-like presentation of parvovirus B19 infection. Am J Med 2001; 111:573-5. [PMID: 11705435 DOI: 10.1016/s0002-9343(01)00904-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
38
|
James JA, Harley JB, Scofield RH. Role of viruses in systemic lupus erythematosus and Sjögren syndrome. Curr Opin Rheumatol 2001; 13:370-6. [PMID: 11604590 DOI: 10.1097/00002281-200109000-00005] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Systemic lupus erythematosus and Sjögren syndrome remain elusive in the description of their underlying etiologic causes and pathogenic mechanisms. Although underlying genetic predisposition appears to contribute to both diseases based on twin and other genetic studies, additional factors must play a role. Over the decades additional factors, such as hormonal influence, UV light, environmental exposures (e.g., silica, solvents), and infectious agents have been postulated to play a role. Over the past few years additional information has been published concerning roles of various infectious agents in both lupus and Sjögren syndrome. Although the understanding of this field is still incomplete, significant advances are being made.
Collapse
Affiliation(s)
- J A James
- Arthritis and Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma 73104, USA.
| | | | | |
Collapse
|
39
|
Lukashov VV, Goudsmit J. Evolutionary relationships among parvoviruses: virus-host coevolution among autonomous primate parvoviruses and links between adeno-associated and avian parvoviruses. J Virol 2001; 75:2729-40. [PMID: 11222696 PMCID: PMC115897 DOI: 10.1128/jvi.75.6.2729-2740.2001] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The current classification of parvoviruses is based on virus host range and helper virus dependence, while little data on evolutionary relationships among viruses are available. We identified and analyzed 472 sequences of parvoviruses, among which there were (virtually) full-length genomes of all 41 viruses currently recognized as individual species within the family Parvoviridae. Our phylogenetic analysis of full-length genomes as well as open reading frames distinguished three evolutionary groups of parvoviruses from vertebrates: (i) the human helper-dependent adeno-associated virus (AAV) serotypes 1 to 6 and the autonomous avian parvoviruses; (ii) the bovine, chipmunk, and autonomous primate parvoviruses, including human viruses B19 and V9; and (iii) the parvoviruses from rodents (except for chipmunks), carnivores, and pigs. Each of these three evolutionary groups could be further subdivided, reflecting both virus-host coevolution and multiple cross-species transmissions in the evolutionary history of parvoviruses. No parvoviruses from invertebrates clustered with vertebrate parvoviruses. Our analysis provided evidence for negative selection among parvoviruses, the independent evolution of their genes, and recombination among parvoviruses from rodents. The topology of the phylogenetic tree of autonomous human and simian parvoviruses matched exactly the topology of the primate family tree, as based on the analysis of primate mitochondrial DNA. Viruses belonging to the AAV group were not evolutionarily linked to other primate parvoviruses but were linked to the parvoviruses of birds. The two lineages of human parvoviruses may have resulted from independent ancient zoonotic infections. Our results provide an argument for reclassification of Parvovirinae based on evolutionary relationships among viruses.
Collapse
Affiliation(s)
- V V Lukashov
- Department of Human Retrovirology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands.
| | | |
Collapse
|
40
|
Scheurlen W, Ramasubbu K, Wachowski O, Hemauer A, Modrow S. Chronic autoimmune thrombopenia/neutropenia in a boy with persistent parvovirus B19 infection. J Clin Virol 2001; 20:173-8. [PMID: 11166667 DOI: 10.1016/s1386-6532(00)00179-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE We report an 11-year-old boy presenting with splenomegaly, chronic thrombocytopenia and concordant neutropenia. RESULTS In contrast to autoantibodies against platelets, there were no detectable neutrophil-specific autoantibodies present in this patient. Extensive serologic investigations revealed increased IgM- and IgG-antibody titers against parvovirus B19. A nested polymerase chain reaction (PCR) showed parvovirus B19-specific sequences in the patient's bone-marrow cells but not in the serum. Specific antibodies against the structural proteins VP1 and VP2 in addition to those against non-structural protein NS1 of parvovirus B19 were detected by Western blot analysis. Thrombocytopenia and neutropenia responded to immunosuppressive therapy and subsequent splenectomy, the latter being necessary due to severe side-effects of steroid medication. CONCLUSION Autoimmune thrombocytopenia/neutropenia may have been triggered and/or sustained by a chronic parvovirus B19 infection. Patients with this very rare disorder should be screened for this virus.
Collapse
Affiliation(s)
- W Scheurlen
- Universitäts-Kinderklinik Mannheim, Fakultät für Klinische Medizin Mannheim der Universität Heidelberg, Theodor-Kutzer-Ufer 1-3, D-68167 Mannheim, Germany.
| | | | | | | | | |
Collapse
|
41
|
Abstract
Aches and pains in children can arise from multiple problems, varying from a reaction to minor intercurrent infection that rapidly improves to the presence of severe skeletal lesions such as malignancy; they can also be part of a skeletal dysplasia. All cases require a good history (including family history), a full examination, and basic blood tests, which include the erythrocyte sedimentation rate, hemoglobin, white count, platelets, rheumatoid factor, and antinuclear factor. Other tests need be performed only when suspicion has been aroused. Recognition of unusual syndromes is important; no child should be labeled as having juvenile idiopathic arthritis unless there is a clear history with the presence of soft tissue swelling in appropriate sites and other causes for joint pain have been excluded. The conditions that most frequently mimic systemic onset juvenile arthritis are infections, which may have been partially treated, inflammatory bowel disease, malignancy, familial Mediterranean Fever, and the rarer connective tissue diseases, in particular systemic lupus erythematosus. Bacterial infection should be suspected in a child who is feverish and toxic, with a single hot swollen joint that has limited movement and is often rigidly guarded. Should such a child have already received antibiotics, general symptoms may well be minimal, so one is left with the history and a swollen and painful joint. Aspiration for investigation of the synovial fluid as well as blood tests should be undertaken immediately to establish the nature of any underlying infection.
Collapse
|
42
|
Affiliation(s)
- J R Kerr
- Department of Medical Microbiology and Virology, Manchester Royal Infirmary, Oxford Road, Manchester M13 9WL, UK.
| |
Collapse
|
43
|
Hemauer A, Gigler A, Searle K, Beckenlehner K, Raab U, Broliden K, Wolf H, Enders G, Modrow S. Seroprevalence of parvovirus B19 NS1-specific IgG in B19-infected and uninfected individuals and in infected pregnant women. J Med Virol 2000; 60:48-55. [PMID: 10568763 DOI: 10.1002/(sici)1096-9071(200001)60:1<48::aid-jmv9>3.0.co;2-e] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Parvovirus B19 is the causative agent of erythema infectiosum in children, but the virus is associated with an increasing range of different diseases. These include acute and chronic arthritis, hydrops fetalis in pregnant women, aplastic anemia, and thrombocytopenia. The host's immune response is directed against the viral structural proteins VP1 and VP2. This study investigated the presence of IgG against the viral nonstructural protein NS1 using Western blot. Serum panels from healthy individuals, B19-infected pregnant women, and various disease groups were tested. The disease groups included patients with symptoms that may be linked to parvovirus B19 infection. The results showed that IgG against the NS1 protein was present in 22% of healthy individuals with past B19 infection. In cases of persistent or prolonged B19 infections, the prevalence of NS1-specific antibodies was as high as 80%. It is concluded that NS1-specific IgG may be used as an indicator of chronic or more severe courses of parvovirus B19 infections.
Collapse
Affiliation(s)
- A Hemauer
- Institut für Medizinische Mikrobiologie, Universität Regensburg, Regensburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|