1
|
Pedreañez A, Carrero Y, Vargas R, Hernandez-Fonseca JP, Hernandez-Fonseca H, Mosquera JA. Role of Gut Microbiota in Dengue. Rev Med Virol 2024; 34:e2577. [PMID: 39215460 DOI: 10.1002/rmv.2577] [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: 04/28/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Dengue is a disease caused by a flavivirus (DENV) and transmitted by the bite of a mosquito, primarily the Aedes aegypti and Aedes albopictus species. Previous studies have demonstrated a relationship between the host gut microbiota and the evolution of dengue. It seems to be a bidirectional relationship, in which the DENV can affect the microbiota by inducing alterations related to intestinal permeability, leading to the release of molecules from microbiota dysbiosis that can influence the evolution of dengue. The role of angiotensin II (Ang II) in the microbiota/dengue relationship is not well understood, but it is known that the renin-angiotensin system (RAS) is present in the intestinal tract and interacts with the gut microbiota. The possible effect of Ang II on the microbiota/Ang II/dengue relationship can be summarised as follows: the presence of Ang II induced hypertension, the increase in angiotensinogen, chymase, and microRNAs during the disease, the induction of vascular dysfunction, the production of trimethylamine N-oxide and the brain/microbiota relationship, all of which are elements present in dengue that could be part of the microbiota/Ang II/dengue interactions. These findings suggest the potential use of Ang II synthesis blockers and the use of AT1 receptor antagonists as therapeutic drugs in dengue.
Collapse
Affiliation(s)
- Adriana Pedreañez
- Cátedra de Inmunología, Escuela de Bioanálisis, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Yenddy Carrero
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Renata Vargas
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Juan P Hernandez-Fonseca
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
- Servicio de Microscopia Electrónica, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, España
| | - Hugo Hernandez-Fonseca
- Facultad de Ciencias Veterinarias, Universidad del Zulia, Maracaibo, Venezuela
- Anatomy, Physiology and Pharmacology Department, School of Veterinary Medicine, Saint George's University, Saint George, Grenada
| | - Jesús A Mosquera
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| |
Collapse
|
2
|
Weber WC, Andoh TF, Kreklywich CN, Streblow ZJ, Denton M, Streblow MM, Powers JM, Sulgey G, Medica S, Dmitriev I, Curiel DT, Haese NN, Streblow DN. Nonreciprocity in CHIKV and MAYV Vaccine-Elicited Protection. Vaccines (Basel) 2024; 12:970. [PMID: 39340002 PMCID: PMC11435824 DOI: 10.3390/vaccines12090970] [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: 07/19/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Chikungunya virus (CHIKV) is a pathogenic arthritogenic alphavirus responsible for large-scale human epidemics for which a vaccine was recently approved for use. Mayaro virus (MAYV) is a related emerging alphavirus with epidemic potential with circulation overlap potential with CHIKV. We previously reported the ability of a non-replicating human adenovirus (AdV)-vectored vaccine expressing the MAYV structural polyprotein to protect against disease in mice following challenge with MAYV, CHIKV and UNAV. Herein, we evaluated mouse immunity and protective efficacy for an AdV-CHIKV full structural polyprotein vaccine in combination with heterologous AdV-MAYV prime/boost regimens versus vaccine coadministration. Heterologous prime/boost regimens skewed immunity toward the prime vaccine antigen but allowed for a boost of cross-neutralizing antibodies, while vaccine co-administration elicited robust, balanced responses capable of boosting. All immunization strategies protected against disease from homologous virus infection, but reciprocal protective immunity differences were revealed upon challenge with heterologous viruses. In vivo passive transfer experiments reproduced the inequity in reciprocal cross-protection after heterologous MAYV challenge. We detected in vitro antibody-dependent enhancement of MAYV replication, suggesting a potential mechanism for the lack of cross-protection. Our findings provide important insights into rational alphavirus vaccine design that may have important implications for the evolving alphavirus vaccine landscape.
Collapse
Affiliation(s)
- Whitney C. Weber
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Takeshi F. Andoh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Craig N. Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Zachary J. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Michael Denton
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Magdalene M. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - John M. Powers
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Gauthami Sulgey
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Samuel Medica
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Igor Dmitriev
- Cancer Biology Division, Department of Radiation Oncology, Washington University, St. Louis, MO 63110, USA; (I.D.); (D.T.C.)
| | - David T. Curiel
- Cancer Biology Division, Department of Radiation Oncology, Washington University, St. Louis, MO 63110, USA; (I.D.); (D.T.C.)
| | - Nicole N. Haese
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA; (W.C.W.); (T.F.A.); (C.N.K.); (Z.J.S.); (M.D.); (J.M.P.); (G.S.); (S.M.); (N.N.H.)
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| |
Collapse
|
3
|
Zhang L, Feng X, Chen W, Wang B, He S, Fan H, Liu D. Non-infectious immune complexes downregulate the production of interferons and tumor necrosis factor-α in primary porcine alveolar macrophages in vitro. Front Vet Sci 2024; 11:1420466. [PMID: 38962699 PMCID: PMC11221350 DOI: 10.3389/fvets.2024.1420466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) caused by the PRRS virus (PRRSV) has been harming the pig industry worldwide for nearly 40 years. Although scientific researchers have made substantial efforts to explore PRRSV pathogenesis, the immune factors influencing PRRSV infection still need to be better understood. Infectious virus-antibody immune complexes (ICs) formed by PRRSV and sub-or non-neutralizing antibodies specific for PRRSV may significantly promote the development of PRRS by enhancing PRRSV replication through antibody-dependent enhancement. However, nothing is known about whether PRRSV infection is affected by non-infectious ICs (NICs) formed by non-pathogenic/infectious antigens and corresponding specific antibodies. Here, we found that PRRSV significantly induced the transcripts and proteins of interferon-α (IFN-α), IFN-β, IFN-γ, IFN-λ1, and tumor necrosis factor-α (TNF-α) in vitro primary porcine alveolar macrophages (PAMs) in the early stage of infection. Our results showed that NICs formed by rabbit-negative IgG (RNI) and pig anti-RNI specific IgG significantly reduced the transcripts and proteins of IFN-α, IFN-β, IFN-γ, IFN-λ1, and TNF-α in vitro PAMs and significantly elevated the transcripts and proteins of interleukine-10 (IL-10) and transforming growth factor-β1 (TGF-β1) in vitro PAMs. NICs-mediated PRRSV infection showed that NICs not only significantly decreased the induction of IFN-α, IFN-β, IFN-γ, IFN-λ1, and TNF-α by PRRSV but also significantly increased the induction of IL-10 and TGF-β1 by PRRSV and considerably enhanced PRRSV replication in vitro PAMs. Our data suggested that NICs could downregulate the production of antiviral cytokines (IFN-α/β/γ/λ1 and TNF-α) during PRRSV infection in vitro and facilitated PRRSV proliferation in its host cells by inhibiting innate antiviral immune response. This study elucidated one novel immune response to PRRSV infection, which would enhance our understanding of the pathogenesis of PRRSV.
Collapse
Affiliation(s)
| | | | | | | | | | - Hongjie Fan
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Deyi Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| |
Collapse
|
4
|
Sawant J, Patil A, Kurle S. A Review: Understanding Molecular Mechanisms of Antibody-Dependent Enhancement in Viral Infections. Vaccines (Basel) 2023; 11:1240. [PMID: 37515055 PMCID: PMC10384352 DOI: 10.3390/vaccines11071240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Antibody Dependent Enhancement (ADE) of an infection has been of interest in the investigation of many viruses. It is associated with the severity of the infection. ADE is mediated by non-neutralizing antibodies, antibodies at sub-neutralizing concentrations, or cross-reactive non-neutralizing antibodies. Treatments like plasma therapy, B cell immunizations, and antibody therapies may trigger ADE. It is seen as an impediment to vaccine development as well. In viruses including the Dengue virus (DENV), severe acute respiratory syndrome (SARS) virus, Middle East respiratory syndrome (MERS) virus, human immunodeficiency virus (HIV), Ebola virus, Zika virus, and influenza virus, the likely mechanisms of ADE are postulated and described. ADE improves the likelihood of productively infecting cells that are expressing the complement receptor or the Fc receptor (FcR) rather than the viral receptors. ADE occurs when the FcR, particularly the Fc gamma receptor, and/or complement system, particularly Complement 1q (C1q), allow the entry of the virus-antibody complex into the cell. Moreover, ADE alters the innate immune pathways to escape from lysis, promoting viral replication inside the cell that produces viral particles. This review discusses the involvement of FcR and the downstream immunomodulatory pathways in ADE, the complement system, and innate antiviral signaling pathways modification in ADE and its impact on facilitating viral replication. Additionally, we have outlined the modes of ADE in the cases of different viruses reported until now.
Collapse
Affiliation(s)
- Jyoti Sawant
- HIV Drug Resistance Laboratory, ICMR-National AIDS Research Institute, Pune 411026, India
| | - Ajit Patil
- HIV Drug Resistance Laboratory, ICMR-National AIDS Research Institute, Pune 411026, India
| | - Swarali Kurle
- HIV Drug Resistance Laboratory, ICMR-National AIDS Research Institute, Pune 411026, India
| |
Collapse
|
5
|
Aghbash PS, Rasizadeh R, Arefi V, Nahand JS, Baghi HB. Immune-checkpoint expression in antigen-presenting cells (APCs) of cytomegaloviruses infection after transplantation: as a diagnostic biomarker. Arch Microbiol 2023; 205:280. [PMID: 37430000 DOI: 10.1007/s00203-023-03623-8] [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: 05/25/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/12/2023]
Abstract
Cytomegalovirus (CMV), a member of the Herpesviridae family, mostly causes only slight feverish symptoms or can be asymptomatic in immunocompetent individuals. However, it is known to be particularly a significant cause of morbidity in immunocompromised patients, including transplant recipients, whose immune system has been weakened due to the consumption of immunosuppressor drugs. Therefore, the diagnosis of CMV infection after transplantation is crucial. New diagnostic methods for the quick detection of CMV have been developed as a result of understanding the clinical importance of invasive CMV. Antigen-presenting cells (APCs) and T cells are important components of the immune system and it may be possible to diagnose viral infections using immunological markers, such as lymphocytosis, cytotoxic T lymphocytes (CTL), and serum cytokine levels. Moreover, PD-1, CTLA 4, and TIGIT, which are expressed on certain T cells and antigen-presenting cells, are over-expressed during the infection. The assessment of CMV infection based on T cell and APC activity, and the expression of immunological checkpoints, can be helpful for the diagnosis of transplant patients at risk for CMV infection. In this review, we will investigate how immune checkpoints affect immune cells and how they impair organ transplantation after CMV infection.
Collapse
Affiliation(s)
- Parisa Shiri Aghbash
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reyhaneh Rasizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Arefi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
- Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran
| | - Hossein Bannazadeh Baghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, 5166/15731, Iran.
| |
Collapse
|
6
|
Mosquera-Sulbaran JA, Pedreañez A, Hernandez-Fonseca JP, Hernandez-Fonseca H. Angiotensin II and dengue. Arch Virol 2023; 168:191. [PMID: 37368044 DOI: 10.1007/s00705-023-05814-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 05/09/2023] [Indexed: 06/28/2023]
Abstract
Dengue is a disease caused by a flavivirus that is transmitted principally by the bite of an Aedes aegypti mosquito and represents a major public-health problem. Many studies have been carried out to identify soluble factors that are involved in the pathogenesis of this infection. Cytokines, soluble factors, and oxidative stress have been reported to be involved in the development of severe disease. Angiotensin II (Ang II) is a hormone with the ability to induce the production of cytokines and soluble factors related to the inflammatory processes and coagulation disorders observed in dengue. However, a direct involvement of Ang II in this disease has not been demonstrated. This review primarily summarizes the pathophysiology of dengue, the role of Ang II in various diseases, and reports that are highly suggestive of the involvement of this hormone in dengue.
Collapse
Affiliation(s)
- Jesus A Mosquera-Sulbaran
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, 4001-A, Venezuela.
| | - Adriana Pedreañez
- Cátedra de Inmunología, Escuela de Bioanálisis, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Juan Pablo Hernandez-Fonseca
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette", Facultad de Medicina, Universidad del Zulia, Maracaibo, 4001-A, Venezuela
- Servicio de Microscopia Electronica del Centro Nacional de Biotecnologia (CNB- CSIC) Madrid, Madrid, España
| | - Hugo Hernandez-Fonseca
- Department of Anatomy, Physiology and Pharmacology, School of Veterinary Medicine, Saint George's University, True Blue, West Indies, Grenada
| |
Collapse
|
7
|
Guerrero-Arguero I, Tellez-Freitas CM, Weber KS, Berges BK, Robison RA, Pickett BE. Alphaviruses: Host pathogenesis, immune response, and vaccine & treatment updates. J Gen Virol 2021; 102. [PMID: 34435944 DOI: 10.1099/jgv.0.001644] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Human pathogens belonging to the Alphavirus genus, in the Togaviridae family, are transmitted primarily by mosquitoes. The signs and symptoms associated with these viruses include fever and polyarthralgia, defined as joint pain and inflammation, as well as encephalitis. In the last decade, our understanding of the interactions between members of the alphavirus genus and the human host has increased due to the re-appearance of the chikungunya virus (CHIKV) in Asia and Europe, as well as its emergence in the Americas. Alphaviruses affect host immunity through cytokines and the interferon response. Understanding alphavirus interactions with both the innate immune system as well as the various cells in the adaptive immune systems is critical to developing effective therapeutics. In this review, we summarize the latest research on alphavirus-host cell interactions, underlying infection mechanisms, and possible treatments.
Collapse
Affiliation(s)
- Israel Guerrero-Arguero
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA.,Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - K Scott Weber
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Bradford K Berges
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Richard A Robison
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Brett E Pickett
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| |
Collapse
|
8
|
Yang B, Yang KD. Immunopathogenesis of Different Emerging Viral Infections: Evasion, Fatal Mechanism, and Prevention. Front Immunol 2021; 12:690976. [PMID: 34335596 PMCID: PMC8320726 DOI: 10.3389/fimmu.2021.690976] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/14/2021] [Indexed: 12/16/2022] Open
Abstract
Different emerging viral infections may emerge in different regions of the world and pose a global pandemic threat with high fatality. Clarification of the immunopathogenesis of different emerging viral infections can provide a plan for the crisis management and prevention of emerging infections. This perspective article describes how an emerging viral infection evolves from microbial mutation, zoonotic and/or vector-borne transmission that progresses to a fatal infection due to overt viremia, tissue-specific cytotropic damage or/and immunopathology. We classified immunopathogenesis of common emerging viral infections into 4 categories: 1) deficient immunity with disseminated viremia (e.g., Ebola); 2) pneumocytotropism with/without later hyperinflammation (e.g., COVID-19); 3) augmented immunopathology (e.g., Hanta); and 4) antibody-dependent enhancement of infection with altered immunity (e.g., Dengue). A practical guide to early blocking of viral evasion, limiting viral load and identifying the fatal mechanism of an emerging viral infection is provided to prevent and reduce the transmission, and to do rapid diagnoses followed by the early treatment of virus neutralization for reduction of morbidity and mortality of an emerging viral infection such as COVID-19.
Collapse
Affiliation(s)
- Betsy Yang
- Department of Medicine, Kaiser Permanente Oakland Medical Center, Oakland, CA, United States
| | - Kuender D. Yang
- DIvision of Medical Research, Mackay Children’s Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang Ming University, Taipei, Taiwan
- Department of Microbiology & Immunology, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
9
|
Lidbury BA. Ross River Virus Immune Evasion Strategies and the Relevance to Post-viral Fatigue, and Myalgic Encephalomyelitis Onset. Front Med (Lausanne) 2021; 8:662513. [PMID: 33842517 PMCID: PMC8024622 DOI: 10.3389/fmed.2021.662513] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 02/24/2021] [Indexed: 01/06/2023] Open
Abstract
Ross River virus (RRV) is an endemic Australian arbovirus, and member of the Alphavirus family that also includes Chikungunya virus (CHIK). RRV is responsible for the highest prevalence of human disease cases associated with mosquito-borne transmission in Australia, and has long been a leading suspect in cases of post-viral fatigue syndromes, with extrapolation of this link to Myalgic Encephalomyelitis (ME). Research into RRV pathogenesis has revealed a number of immune evasion strategies, impressive for a virus with a genome size of 12 kb (plus strand RNA), which resonate with insights into viral pathogenesis broadly. Drawing from observations on RRV immune evasion, mechanisms of relevance to long term idiopathic fatigue are featured as a perspective on infection and eventual ME symptoms, which include considerations of; (1) selective pro-inflammatory gene suppression post antibody-dependent enhancement (ADE) of RRV infection, (2) Evidence from other virus families of immune disruption and evasion post-ADE, and (3) how virally-driven immune evasion may impact on mitochondrial function via target of rapamycin (TOR) complexes. In light of these RRV measures to counter the host immune - inflammatory responses, links to recent discoveries explaining cellular, immune and metabolomic markers of ME will be explored and discussed, with the implications for long-COVID post SARS-CoV-2 also considered. Compelling issues on the connections between virally-induced alterations in cytokine expression, for example, will be of particular interest in light of energy pathways, and how these perturbations manifest clinically.
Collapse
Affiliation(s)
- Brett A Lidbury
- National Centre for Epidemiology and Population Health, Research School of Population Health, The Australian National University, Canberra, ACT, Australia
| |
Collapse
|
10
|
Wen J, Cheng Y, Ling R, Dai Y, Huang B, Huang W, Zhang S, Jiang Y. Antibody-dependent enhancement of coronavirus. Int J Infect Dis 2020; 100:483-489. [PMID: 32920233 PMCID: PMC7483033 DOI: 10.1016/j.ijid.2020.09.015] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 07/28/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022] Open
Abstract
Antibody-dependent enhancement (ADE) exists in several kinds of virus. It has a negative influence on antibody therapy for viral infection. This effect was first identified in dengue virus and has since also been described for coronavirus. To date, the rapid spread of the newly emerged coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), causing coronavirus disease 2019 (COVID-19), has affected over 3.8 million people across the globe. The novel coronavirus poses a great challenge and has caused a wave of panic. In this review, antibody-dependent enhancements in dengue virus and two kinds of coronavirus are summarized. Possible solutions for the effects are reported. We also speculate that ADE may exist in SARS-CoV-2.
Collapse
Affiliation(s)
- Jieqi Wen
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Yifan Cheng
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Rongsong Ling
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Yarong Dai
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Boxuan Huang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Wenjie Huang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Siyan Zhang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| | - Yizhou Jiang
- Institute for Advanced Study, Shenzhen University, Shenzhen 518067, China.
| |
Collapse
|
11
|
Antibody-Mediated Porcine Reproductive and Respiratory Syndrome Virus Infection Downregulates the Production of Interferon-α and Tumor Necrosis Factor-α in Porcine Alveolar Macrophages via Fc Gamma Receptor I and III. Viruses 2020; 12:v12020187. [PMID: 32046249 PMCID: PMC7077232 DOI: 10.3390/v12020187] [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: 01/15/2020] [Revised: 02/01/2020] [Accepted: 02/06/2020] [Indexed: 12/24/2022] Open
Abstract
Antibody-dependent enhancement (ADE) contributes to the pathogenesis of porcine reproductive and respiratory syndrome virus (PRRSV)-persistent infection. However, the mechanisms of PRRSV-ADE infection are still confusing. A clear understanding of the event upon virus infection by the ADE pathway has become crucial for developing efficient intervention of the PRRSV infection. In this study, an ADE assay showed that PRRSV-ADE infection in porcine alveolar macrophages (AMs) significantly decreased the production of interferon-α (IFN-α) and tumor necrosis factor-α (TNF-α), and significantly increased the production of interleukine-10 (IL-10). A gene knockdown assay based on small interfering RNA (siRNA) showed that both Fc gamma receptor I (FcγRI) and FcγRIII in porcine AMs were involved in PRRSV-ADE infection. An activation assay showed that specific activation of FcγRI or FcγRIII in porcine AMs during PRRSV infection not only significantly decreased the production of IFN-α and TNF-α, but also significantly increased the production of IL-10 and significantly facilitated PRRSV replication. In conclusion, our studies suggested that ADE downregulated the production of IFN-α and TNF-α in porcine AMs maybe via FcγRI and FcγRIII, thereby leading to enhanced PRRSV infection.
Collapse
|
12
|
Effects of Chikungunya virus immunity on Mayaro virus disease and epidemic potential. Sci Rep 2019; 9:20399. [PMID: 31892710 PMCID: PMC6938517 DOI: 10.1038/s41598-019-56551-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/13/2019] [Indexed: 12/30/2022] Open
Abstract
Mayaro virus (MAYV) causes an acute febrile illness similar to that produced by chikungunya virus (CHIKV), an evolutionary relative in the Semliki Forest virus complex of alphaviruses. MAYV emergence is typically sporadic, but recent isolations and outbreaks indicate that the virus remains a public health concern. Given the close phylogenetic and antigenic relationship between CHIKV and MAYV, and widespread distribution of CHIKV, we hypothesized that prior CHIKV immunity may affect MAYV pathogenesis and/or influence its emergence potential. We pre-exposed immunocompetent C57BL/6 and immunocompromised A129 or IFNAR mice to wild-type CHIKV, two CHIKV vaccines, or a live-attenuated MAYV vaccine, and challenged with MAYV. We observed strong cross-protection against MAYV for mice pre-exposed to wild-type CHIKV, and moderately but significantly reduced cross-protection from CHIKV-vaccinated animals. Immunity to other alphavirus or flavivirus controls provided no protection against MAYV disease or viremia. Mechanistic studies suggested that neutralizing antibodies alone can mediate this protection, with T-cells having no significant effect on diminishing disease. Finally, human sera obtained from naturally acquired CHIKV infection cross-neutralized MAYV at high titers in vitro. Altogether, our data suggest that CHIKV infection can confer cross-protective effects against MAYV, and the resultant reduction in viremia may limit the emergence potential of MAYV.
Collapse
|
13
|
Ullm F, Riedl P, Machado de Amorim A, Patzschke A, Weiß R, Hauschildt S, Franke K, Anderegg U, Pompe T. 3D Scaffold-Based Macrophage Fibroblast Coculture Model Reveals IL-10 Dependence of Wound Resolution Phase. ACTA ACUST UNITED AC 2019; 4:e1900220. [PMID: 32293120 DOI: 10.1002/adbi.201900220] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/08/2019] [Indexed: 12/18/2022]
Abstract
Persistent inflammation and impaired repair in dermal wound healing are frequently associated with cell-cell and cell-matrix miscommunication. A direct coculture model of primary human myofibroblasts (MyoFB) and M-CSF-differentiated macrophages (M-Mɸ) in fibrillar three-dimensional Collagen I (Coll I) matrices is developed to study intercellular interactions. The coculture experiments reveal the number of M-Mɸ regulated MyoFB dedifferentiation in a dose-dependent manner. The amount of MyoFB decreases in dependence of the number of cocultured M-Mɸ, even in the presence of MyoFB-inducing transforming growth factor β1 (TGF-β1 ). Gene expression analysis of matrix proteins (collagen I, collagen III, ED-A-fibronectin) confirms the results of an altered MyoFB phenotype. Additionally, M-Mɸ is shown to be the main source of secreted cytokine interleukin-10 (IL-10), which is suggested to affect MyoFB dedifferentiation. These findings indicate a paracrine impact of IL-10 secretion by M-Mɸ on the MyoFB differentiation status counteracting the TGF-β1 -driven MyoFB activation. Hence, the in vitro coculture model simulates physiological situations during wound resolution and underlines the importance of paracrine IL-10 signals by M-Mɸ. In sum, the 3D Coll I-based matrices with a MyoFB-M-Mɸ coculture form a highly relevant biomimetic model of late stages of wound healing.
Collapse
Affiliation(s)
- Franziska Ullm
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, 04103, Leipzig, Germany
| | - Philipp Riedl
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, 04103, Leipzig, Germany
| | | | - Aline Patzschke
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, 04103, Leipzig, Germany
| | - Ronald Weiß
- Institute of Clinical Immunology, Medical Faculty, Leipzig University, 04103, Leipzig, Germany
| | - Sunna Hauschildt
- Institute of Biology, Faculty of Life Sciences, Leipzig University, 04103, Leipzig, Germany
| | - Katja Franke
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, 04103, Leipzig, Germany
| | - Ulf Anderegg
- Department of Dermatology, Venerology and Allergology, Medical Faculty, Leipzig University Leipzig, 04103, Leipzig, Germany
| | - Tilo Pompe
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, 04103, Leipzig, Germany
| |
Collapse
|
14
|
Yu S, Su C, Luo X. Impact of infection on transplantation tolerance. Immunol Rev 2019; 292:243-263. [PMID: 31538351 PMCID: PMC6961566 DOI: 10.1111/imr.12803] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022]
Abstract
Allograft tolerance is the ultimate goal of organ transplantation. Current strategies for tolerance induction mainly focus on inhibiting alloreactive T cells while promoting regulatory immune cells. Pathogenic infections may have direct impact on both effector and regulatory cell populations, therefore can alter host susceptibility to transplantation tolerance induction as well as impair the quality and stability of tolerance once induced. In this review, we will discuss existing data demonstrating the effect of infections on transplantation tolerance, with particular emphasis on the role of the stage of infection (acute, chronic, or latent) and the stage of tolerance (induction or maintenance) in this infection-tolerance interaction. While the deleterious effect of acute infection on tolerance is mainly driven by proinflammatory cytokines induced shortly after the infection, chronic infection may generate exhausted T cells that could in fact facilitate transplantation tolerance. In addition to pathogenic infections, commensal intestinal microbiota also has numerous significant immunomodulatory effects that can shape the host alloimmunity following transplantation. A comprehensive understanding of these mechanisms is crucial for the development of therapeutic strategies for robustly inducing and stably maintaining transplantation tolerance while preserving host anti-pathogen immunity in clinically relevant scenarios.
Collapse
Affiliation(s)
- Shuangjin Yu
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
- Division of Organ transplantation, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Chang Su
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, United States
- Duke Transplant Center, Duke University School of Medicine, Durham, NC 27710, United States
| |
Collapse
|
15
|
Smatti MK, Al Thani AA, Yassine HM. Viral-Induced Enhanced Disease Illness. Front Microbiol 2018; 9:2991. [PMID: 30568643 PMCID: PMC6290032 DOI: 10.3389/fmicb.2018.02991] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/19/2018] [Indexed: 12/24/2022] Open
Abstract
Understanding immune responses to viral infections is crucial to progress in the quest for effective infection prevention and control. The host immunity involves various mechanisms to combat viral infections. Under certain circumstances, a viral infection or vaccination may result in a subverted immune system, which may lead to an exacerbated illness. Clinical evidence of enhanced illness by preexisting antibodies from vaccination, infection or maternal passive immunity is available for several viruses and is presumptively proposed for other viruses. Multiple mechanisms have been proposed to explain this phenomenon. It has been confirmed that certain infection- and/or vaccine-induced immunity could exacerbate viral infectivity in Fc receptor- or complement bearing cells- mediated mechanisms. Considering that antibody dependent enhancement (ADE) is a major obstacle in vaccine development, there are continues efforts to understand the underlying mechanisms through identification of the epitopes and antibodies responsible for disease enhancement or protection. This review discusses the recent findings on virally induced ADE, and highlights the potential mechanisms leading to this condition.
Collapse
Affiliation(s)
- Maria K Smatti
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | - Hadi M Yassine
- Biomedical Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
16
|
Rasa S, Nora-Krukle Z, Henning N, Eliassen E, Shikova E, Harrer T, Scheibenbogen C, Murovska M, Prusty BK. Chronic viral infections in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS). J Transl Med 2018; 16:268. [PMID: 30285773 PMCID: PMC6167797 DOI: 10.1186/s12967-018-1644-y] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 09/24/2018] [Indexed: 12/15/2022] Open
Abstract
Background and main text Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a complex and controversial clinical condition without having established causative factors. Increasing numbers of cases during past decade have created awareness among patients as well as healthcare professionals. Chronic viral infection as a cause of ME/CFS has long been debated. However, lack of large studies involving well-designed patient groups and validated experimental set ups have hindered our knowledge about this disease. Moreover, recent developments regarding molecular mechanism of pathogenesis of various infectious agents cast doubts over validity of several of the past studies. Conclusions This review aims to compile all the studies done so far to investigate various viral agents that could be associated with ME/CFS. Furthermore, we suggest strategies to better design future studies on the role of viral infections in ME/CFS.
Collapse
Affiliation(s)
- Santa Rasa
- Institute of Microbiology and Virology, Rīga Stradiņš University, Riga, Latvia
| | - Zaiga Nora-Krukle
- Institute of Microbiology and Virology, Rīga Stradiņš University, Riga, Latvia
| | - Nina Henning
- Biocenter, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| | - Eva Eliassen
- Biocenter, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| | - Evelina Shikova
- Department of Virology, National Center of Infectious and Parasitic Diseases, Sofia, Bulgaria
| | - Thomas Harrer
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Carmen Scheibenbogen
- Institute for Medical Immunology, Charité-Universitätsmedizin Berlin, Campus Virchow, Berlin, Germany
| | - Modra Murovska
- Institute of Microbiology and Virology, Rīga Stradiņš University, Riga, Latvia
| | - Bhupesh K Prusty
- Biocenter, Chair of Microbiology, University of Würzburg, Würzburg, Germany. .,Institute for Virology and Immunobiology, Würzburg, Germany.
| | | |
Collapse
|
17
|
Antibody-mediated enhancement aggravates chikungunya virus infection and disease severity. Sci Rep 2018; 8:1860. [PMID: 29382880 PMCID: PMC5789897 DOI: 10.1038/s41598-018-20305-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 01/17/2018] [Indexed: 01/18/2023] Open
Abstract
The arthropod-transmitted chikungunya virus (CHIKV) causes a flu-like disease that is characterized by incapacitating arthralgia. The re-emergence of CHIKV and the continual risk of new epidemics have reignited research in CHIKV pathogenesis. Virus-specific antibodies have been shown to control virus clearance, but antibodies present at sub-neutralizing concentrations can also augment virus infection that exacerbates disease severity. To explore this occurrence, CHIKV infection was investigated in the presence of CHIKV-specific antibodies in both primary human cells and a murine macrophage cell line, RAW264.7. Enhanced attachment of CHIKV to the primary human monocytes and B cells was observed while increased viral replication was detected in RAW264.7 cells. Blocking of specific Fc receptors (FcγRs) led to the abrogation of these observations. Furthermore, experimental infection in adult mice showed that animals had higher viral RNA loads and endured more severe joint inflammation in the presence of sub-neutralizing concentrations of CHIKV-specific antibodies. In addition, CHIKV infection in 11 days old mice under enhancing condition resulted in higher muscles viral RNA load detected and death. These observations provide the first evidence of antibody-mediated enhancement in CHIKV infection and pathogenesis and could also be relevant for other important arboviruses such as Zika virus.
Collapse
|
18
|
Cathelicidin-WA polarizes E. coli K88-induced M1 macrophage to M2-like macrophage in RAW264.7 cells. Int Immunopharmacol 2017; 54:52-59. [PMID: 29101873 DOI: 10.1016/j.intimp.2017.10.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/24/2017] [Accepted: 10/11/2017] [Indexed: 12/19/2022]
Abstract
Immune cells - macrophages induced by E. coli K88 will lead to a pro-inflammatory response, which is important in host defense. Cathelicidin-WA (CWA) is an efficient antimicrobial peptide (AMP) and can exert immunomodulatory properties. Many studies have demonstrated that AMP can modulate cellular subsets but whether CWA can regulate macrophage polarization by transferring E. coli K88-induced M1 macrophage towards M2 one that of anti-inflammation remains unclear. In this study, E. coli K88 increased the expression of pro-inflammatory cytokines interleukin-6, interleukin-1β, tumor necrosis factor-α and chemokine CCL3 in RAW264.7 cells with a time-dependent manner, as well as the expression of reactive oxygen species (ROS) and inducible nitric oxide synthase (iNOS). On this basis, CWA significantly decreased the pro-inflammatory molecules but increased the anti-inflammatory mediators interleukin-4, interleukin-10 and other M2-related genes in E. coli K88-induced macrophages. Western blot analysis indicated that CWA suppressed the expression of TLR-4 and the phosphorylation of STAT1 and NF-κB which modulated M1 macrophage while induced the phosphorylation of STAT6 which activated M2 macrophage. Double staining of M1-specific CD86 and M2-specific CD206 also proved the hypothesis. These results suggested that CWA might dampen the inflammation by modulating M1 phenotype to M2 phenotype in E. coli K88-induced macrophages.
Collapse
|
19
|
Taylor A, Foo SS, Bruzzone R, Dinh LV, King NJC, Mahalingam S. Fc receptors in antibody-dependent enhancement of viral infections. Immunol Rev 2016; 268:340-64. [PMID: 26497532 PMCID: PMC7165974 DOI: 10.1111/imr.12367] [Citation(s) in RCA: 188] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sensitization of the humoral immune response to invading viruses and production of antiviral antibodies forms part of the host antiviral repertoire. Paradoxically, for a number of viral pathogens, under certain conditions, antibodies provide an attractive means of enhanced virus entry and replication in a number of cell types. Known as antibody‐dependent enhancement (ADE) of infection, the phenomenon occurs when virus‐antibody immunocomplexes interact with cells bearing complement or Fc receptors, promoting internalization of the virus and increasing infection. Frequently associated with exacerbation of viral disease, ADE of infection presents a major obstacle to the prevention of viral disease by vaccination and is thought to be partly responsible for the adverse effects of novel antiviral therapeutics such as intravenous immunoglobulins. There is a growing body of work examining the intracellular signaling pathways and epitopes responsible for mediating ADE, with a view to aiding rational design of antiviral strategies. With in vitro studies also confirming ADE as a feature of infection for a growing number of viruses, challenges remain in understanding the multilayered molecular mechanisms of ADE and its effect on viral pathogenesis.
Collapse
Affiliation(s)
- Adam Taylor
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, Qld, Australia
| | - Suan-Sin Foo
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, Qld, Australia
| | - Roberto Bruzzone
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong SAR, Hong Kong.,Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | - Luan Vu Dinh
- Discipline of Pathology, Bosch Institute, School of Medical Sciences, Sydney Medical School, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Nicholas J C King
- Discipline of Pathology, Bosch Institute, School of Medical Sciences, Sydney Medical School, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Suresh Mahalingam
- Emerging Viruses and Inflammation Research Group, Institute for Glycomics, Griffith University, Gold Coast, Qld, Australia
| |
Collapse
|
20
|
Yi H, Jiang D, Zhang L, Xiong H, Han F, Wang Y. Developmental expression of STATs, nuclear factor-κB and inflammatory genes in the jejunum of piglets during weaning. Int Immunopharmacol 2016; 36:199-204. [PMID: 27160867 DOI: 10.1016/j.intimp.2016.04.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 04/15/2016] [Accepted: 04/20/2016] [Indexed: 01/05/2023]
Abstract
The signal transducer and activator of transcription (STAT) proteins play essential roles in apoptosis, proliferation and survival. However, the role of STATs in intestinal inflammation during weaning is unclear. This study aimed to investigate developmental expression of STATs, nuclear factor-κB (NF-κB) and inflammatory genes in the jejunum of piglets during weaning. Thirty-two piglets were weaned at 21d and sacrificed at 0, 1, 7, or 14d (n=8) after weaning. Villus height and the villus height/crypt depth ratio were decreased, whereas crypt depth was increased in the jejunum at 7 and 14d after weaning. In addition, the mRNA levels of interferon-γ (IFN-γ), inducible nitric oxide synthase (iNOS), IL-6, IL-8, IL-12 and IL-22 were increased in the jejunum at 7 and 14d after weaning, whereas transforming growth factor-β (TGF-β), suppressor of cytokine signaling 3 (SCOS3) and arginase-1 was decreased. Neutrophil infiltration was increased in the mucosa of the jejunum after weaning. Moreover, phosphorylation of IκB-α, NF-κB, AKT and STAT-3 was increased. However, the phosphorylation of STAT-1 (at 7 and 14d) and STAT-6 (at 1 and 7d) was suppressed in the jejunum after weaning. Treatment of porcine jejunal epithelial (IPEC-J2) cells with the STAT inhibitors fludarabine, niclosamide and teriflunomide, which inhibit the phosphorylation of STAT-1, STAT-3 and STAT-6, respectively, weakened the defense capacity of these cells against bacterial infection. In conclusion, weaning caused severe inflammation associated with activation of the NF-κB and STAT-3 pathways and suppression of STAT-1 and STAT-6 in the jejunum of piglets.
Collapse
Affiliation(s)
- Hongbo Yi
- Laboratory of Animal Nutrition and Feed Science, Department of Animal Science, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Denghu Jiang
- Laboratory of Animal Nutrition and Feed Science, Department of Animal Science, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Lin Zhang
- Laboratory of Animal Nutrition and Feed Science, Department of Animal Science, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Haitao Xiong
- Laboratory of Animal Nutrition and Feed Science, Department of Animal Science, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Feifei Han
- Laboratory of Animal Nutrition and Feed Science, Department of Animal Science, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Yizhen Wang
- Laboratory of Animal Nutrition and Feed Science, Department of Animal Science, Zhejiang University, Hangzhou, Zhejiang Province 310058, China.
| |
Collapse
|
21
|
Huang X, Yue Y, Li D, Zhao Y, Qiu L, Chen J, Pan Y, Xi J, Wang X, Sun Q, Li Q. Antibody-dependent enhancement of dengue virus infection inhibits RLR-mediated Type-I IFN-independent signalling through upregulation of cellular autophagy. Sci Rep 2016; 6:22303. [PMID: 26923481 PMCID: PMC4770412 DOI: 10.1038/srep22303] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/11/2016] [Indexed: 12/25/2022] Open
Abstract
Antibody dependent enhancement (ADE) of dengue virus (DENV) infection is identified as the main risk factor of severe Dengue diseases. Through opsonization by subneutralizing or non-neutralizing antibodies, DENV infection suppresses innate cell immunity to facilitate viral replication. However, it is largely unknown whether suppression of type-I IFN is necessary for a successful ADE infection. Here, we report that both DENV and DENV-ADE infection induce an early ISG (NOS2) expression through RLR-MAVS signalling axis independent of the IFNs signaling. Besides, DENV-ADE suppress this early antiviral response through increased autophagy formation rather than induction of IL-10 secretion. The early induced autophagic proteins ATG5-ATG12 participate in suppression of MAVS mediated ISGs induction. Our findings suggest a mechanism for DENV to evade the early antiviral response before IFN signalling activation. Altogether, these results add knowledge about the complexity of ADE infection and contribute further to research on therapeutic strategies.
Collapse
Affiliation(s)
- Xinwei Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
- Key Laboratory of The Second Affiliated Hospital of Kuming Medical College, Kunming 650101, PR China
| | - Yaofei Yue
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Duo Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Yujiao Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Lijuan Qiu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Junying Chen
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Yue Pan
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Juemin Xi
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Xiaodan Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Qiangming Sun
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Qihan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| |
Collapse
|
22
|
Yi H, Yu C, Zhang H, Song D, Jiang D, Du H, Wang Y. Cathelicidin-BF suppresses intestinal inflammation by inhibiting the nuclear factor-κB signaling pathway and enhancing the phagocytosis of immune cells via STAT-1 in weanling piglets. Int Immunopharmacol 2015; 28:61-9. [DOI: 10.1016/j.intimp.2015.05.034] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 11/16/2022]
|
23
|
Poo YS, Rudd PA, Gardner J, Wilson JAC, Larcher T, Colle MA, Le TT, Nakaya HI, Warrilow D, Allcock R, Bielefeldt-Ohmann H, Schroder WA, Khromykh AA, Lopez JA, Suhrbier A. Multiple immune factors are involved in controlling acute and chronic chikungunya virus infection. PLoS Negl Trop Dis 2014; 8:e3354. [PMID: 25474568 PMCID: PMC4256279 DOI: 10.1371/journal.pntd.0003354] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 10/15/2014] [Indexed: 12/18/2022] Open
Abstract
The recent epidemic of the arthritogenic alphavirus, chikungunya virus (CHIKV) has prompted a quest to understand the correlates of protection against virus and disease in order to inform development of new interventions. Herein we highlight the propensity of CHIKV infections to persist long term, both as persistent, steady-state, viraemias in multiple B cell deficient mouse strains, and as persistent RNA (including negative-strand RNA) in wild-type mice. The knockout mouse studies provided evidence for a role for T cells (but not NK cells) in viraemia suppression, and confirmed the role of T cells in arthritis promotion, with vaccine-induced T cells also shown to be arthritogenic in the absence of antibody responses. However, MHC class II-restricted T cells were not required for production of anti-viral IgG2c responses post CHIKV infection. The anti-viral cytokines, TNF and IFNγ, were persistently elevated in persistently infected B and T cell deficient mice, with adoptive transfer of anti-CHIKV antibodies unable to clear permanently the viraemia from these, or B cell deficient, mice. The NOD background increased viraemia and promoted arthritis, with B, T and NK deficient NOD mice showing high-levels of persistent viraemia and ultimately succumbing to encephalitic disease. In wild-type mice persistent CHIKV RNA and negative strand RNA (detected for up to 100 days post infection) was associated with persistence of cellular infiltrates, CHIKV antigen and stimulation of IFNα/β and T cell responses. These studies highlight that, secondary to antibodies, several factors are involved in virus control, and suggest that chronic arthritic disease is a consequence of persistent, replicating and transcriptionally active CHIKV RNA. The largest epidemic ever recorded for chikungunya virus (CHIKV) started in 2004 in Africa, then spread across Asia and recently caused tens of thousands of cases in Papua New Guinea and the Caribbean. This mosquito-borne alphavirus primarily causes an often debilitating, acute and chronic polyarthritis/polyarthalgia. Despite robust anti-viral immune responses CHIKV is able to persist, with such persistence poorly understood and the likely cause of chronic disease. Herein we highlight the propensity of CHIKV to persist long term, both as a persistent viraemia in different B cell deficient mouse strains, but also as persistent viral RNA in wild-type mice. These studies suggest that, aside from antibodies, other immune factors, such as CD4 T cells and TNF, are active in viraemia control. The work also supports the notion that CHIKV disease, with the exception of encephalitis, is largely an immunopathology. Persistent CHIKV RNA in wild-type mice continues to stimulate type I interferon and T cell responses, with this model of chronic disease recapitulating many of the features seen in chronic CHIKV patients.
Collapse
Affiliation(s)
- Yee Suan Poo
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Penny A. Rudd
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Joy Gardner
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
| | - Jane A. C. Wilson
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - Thibaut Larcher
- Institut National de Recherche Agronomique, Unité Mixte de Recherche 703, Oniris, Nantes, France
| | - Marie-Anne Colle
- Institut National de Recherche Agronomique, Unité Mixte de Recherche 703, Oniris, Nantes, France
| | - Thuy T. Le
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
| | - Helder I. Nakaya
- School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - David Warrilow
- Public Health Virology Laboratory, Department of Health, Queensland Government, Brisbane, Queensland, Australia
| | - Richard Allcock
- Lotterywest State Biomedical Facility Genomics, Royal Perth Hospital, Perth, Western Australia, Australia
| | | | - Wayne A. Schroder
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
| | - Alexander A. Khromykh
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
| | - José A. Lopez
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Natural Sciences, Griffith University, Nathan, Australia
| | - Andreas Suhrbier
- QIMR Berghofer Medical Research Institute, and the Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia
- School of Medicine/School of Molecular and Microbial Sciences, University of Queensland, Brisbane, Queensland, Australia
- School of Natural Sciences, Griffith University, Nathan, Australia
- * E-mail:
| |
Collapse
|
24
|
Abstract
In the healthy gastrointestinal tract, homeostasis is an active process that requires a careful balance of host responses to the enteric luminal contents. Intestinal macrophages and dendritic cells (DCs) comprise a unique group of tissue immune cells that are ideally situated at the interface of the host and the enteric luminal environment to appropriately respond to microbes and ingested stimuli. However, intrinsic defects in macrophage and DC function contribute to the pathogenesis of inflammatory bowel diseases, as highlighted by recent genome-wide association studies. Gastrointestinal macrophages and DCs participate in inflammatory bowel disease development through inappropriate responses to enteric microbial stimuli, inefficient clearance of microbes from host tissues, and impaired transition from appropriate proinflammatory responses to anti-inflammatory responses that promote resolution. By understanding how intestinal macrophages and DCs initiate chronic inflammation, new pathogenesis-based therapeutic strategies to treat human inflammatory bowel diseases will be elucidated.
Collapse
|
25
|
Bao D, Wang R, Qiao S, Wan B, Wang Y, Liu M, Shi X, Guo J, Zhang G. Antibody-dependent enhancement of PRRSV infection down-modulates TNF-α and IFN-β transcription in macrophages. Vet Immunol Immunopathol 2013; 156:128-34. [PMID: 24099951 DOI: 10.1016/j.vetimm.2013.09.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 08/08/2013] [Accepted: 09/12/2013] [Indexed: 11/25/2022]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is an infectious disease, resulting in important economic losses in pig farming. Previous studies have shown that Fcγ receptor (FcγR)-mediated entry of infectious PRRSV immune complexes into macrophages plays a pivotal role in the pathogenesis of the disease. This study demonstrates that PRRSV was able to suppress the transcription of key antiviral genes tumor necrosis factor-α (TNF-α) and interferon-β (IFN-β), when infection was via the ADE pathway. Investigation of this infection pathway found that PRRSV suppresses the antiviral genes by disrupting the transcription of the genes coding for the associated transcription factors interferon regulatory factor-1 (IRF-1), interferon regulatory factor-3 (IRF-3) and nuclear factor kappa B (NF-κB). The ADE pathway of infection allows PRRSV to specifically target antiviral genes and alters the innate intracellular immune responses in macrophages. The ADE mechanism described in this study furthers our understanding of pathogenesis following PRRSV infection and is of general relevance to virally induced disease and in relation to antiviral vaccination strategies.
Collapse
Affiliation(s)
- Dengke Bao
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China; Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Richter K, Oxenius A. Non-neutralizing antibodies protect from chronic LCMV infection independently of activating FcγR or complement. Eur J Immunol 2013; 43:2349-60. [PMID: 23749374 DOI: 10.1002/eji.201343566] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 05/22/2013] [Accepted: 06/04/2013] [Indexed: 01/10/2023]
Abstract
Chronic viral infections lead to CD8(+) T cell exhaustion, characterized by impaired cytokine secretion. The presence of the immune-regulatory cytokine IL-10 promotes chronic lymphocytic choriomeningitis virus (LCMV) Clone 13 infection in mice, whereas the absence of IL-10/IL-10R signaling early during infection results in viral clearance and higher percentages and numbers of antiviral, cytokine-producing T cells. However, it is currently unclear which cell populations and effector molecules are crucial to protect against chronic infection. In this study, we demonstrate that antiviral, LCMV-binding, non-neutralizing antibodies are needed, in addition to CD4(+) and CD8(+) T cells, to clear a high-dose LCMV infection in mice, in the absence of IL-10. The interaction between CD4(+) T cells and B cells in B-cell follicles via CD40/CD40L, in addition to class switch and/or somatic hypermutation, is crucial for viral control in the absence of IL-10. Interestingly, transfer of LCMV-binding non-neutralizing antibodies protected recipients from chronic infection. In addition, viral clearance in the absence of IL-10R signaling was independent of activating Fcγ receptors and complement. These data highlight that non-neutralizing antibodies effectively contribute to the control of LCMV infection when present prior to infection, suggesting that the induction of neutralizing antibodies is not implicitly necessary for the generation of successful vaccines.
Collapse
|
27
|
Lecocq M, Detry B, Guisset A, Pilette C. FcαRI-Mediated Inhibition of IL-12 Production and Priming by IFN-γ of Human Monocytes and Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2013; 190:2362-71. [DOI: 10.4049/jimmunol.1201128] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
28
|
Kozakiewicz L, Phuah J, Flynn J, Chan J. The role of B cells and humoral immunity in Mycobacterium tuberculosis infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 783:225-50. [PMID: 23468112 DOI: 10.1007/978-1-4614-6111-1_12] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tuberculosis (TB) remains a serious threat to public health, causing 2 million deaths annually world-wide. The control of TB has been hindered by the requirement of long duration of treatment involving multiple chemotherapeutic agents, the increased susceptibility to Mycobacterium tuberculosis infection in the HIV-infected population, and the development of multi-drug resistant and extensively resistant strains of tubercle bacilli. An efficacious and cost-efficient way to control TB is the development of effective anti-TB vaccines. This measure requires thorough understanding of the immune response to M. tuberculosis. While the role of cell-mediated immunity in the development of protective immune response to the tubercle bacillus has been well established, the role of B cells in this process is not clearly understood. Emerging evidence suggests that B cells and humoral immunity can modulate the immune response to various intracellular pathogens, including M. tuberculosis. These lymphocytes form conspicuous aggregates in the lungs of tuberculous humans, non-human primates, and mice, which display features of germinal center B cells. In murine TB, it has been shown that B cells can regulate the level of granulomatous reaction, cytokine production, and the T cell response. This chapter discusses the potential mechanisms by which specific functions of B cells and humoral immunity can shape the immune response to intracellular pathogens in general, and to M. tuberculosis in particular. Knowledge of the B cell-mediated immune response to M. tuberculosis may lead to the design of novel strategies, including the development of effective vaccines, to better control TB.
Collapse
Affiliation(s)
- Lee Kozakiewicz
- Department of Medicine and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | | | | |
Collapse
|
29
|
Flipse J, Wilschut J, Smit JM. Molecular mechanisms involved in antibody-dependent enhancement of dengue virus infection in humans. Traffic 2012; 14:25-35. [PMID: 22998156 DOI: 10.1111/tra.12012] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 09/19/2012] [Accepted: 09/21/2012] [Indexed: 12/19/2022]
Abstract
Dengue is the most common arthropod-borne viral infection in humans with ∼50 million cases annually worldwide. In recent decades, a steady increase in the number of severe dengue cases has been seen. Severe dengue disease is most often observed in individuals that have pre-existing immunity against heterotypic dengue subtypes and in infants with low levels of maternal dengue antibodies. The generally accepted hypothesis explaining the immunopathogenesis of severe dengue is called antibody-dependent enhancement of dengue infection. Here, circulating antibodies bind to the newly infecting virus but do not neutralize infection. Rather, these antibodies increase the infected cell mass and virus production. Additionally, antiviral responses are diminished allowing massive virus particle production early in infection. The large infected cell mass and the high viral load are prelude for severe disease development. In this review, we discuss what is known about the trafficking of dengue virus in its human host cells, and the signalling pathways activated after virus detection, both in the absence and presence of antibodies against the virus. This review summarizes work that aims to better understand the complex immunopathogenesis of severe dengue disease.
Collapse
Affiliation(s)
- Jacky Flipse
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | |
Collapse
|
30
|
Zhang Y, Zhou Y, Yang Q, Mu C, Duan E, Chen J, Yang M, Xia P, Cui B. Ligation of Fc gamma receptor IIB enhances levels of antiviral cytokine in response to PRRSV infection in vitro. Vet Microbiol 2012; 160:473-80. [PMID: 22771209 DOI: 10.1016/j.vetmic.2012.06.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 06/19/2012] [Indexed: 10/28/2022]
Abstract
PRRSV infection ADE facilitates the attachment and internalization of the virus onto its host cells, such as monocytes and macrophages, through Fc receptor-mediated endocytosis. FcγRIIB is the only inhibitory receptor with a tyrosine-based inhibitory motif (ITIM) in its cytoplasmic tail, where counters the "ITAM triggered" activation signals and down-regulates phagocytosis. However, porcine FcγRIIB's role in the antiviral immune response to PRRSV infection has not been studied. In this study, our results indicated that selective activation of porcine FcγRIIB in PAM cells up-regulated significantly mRNA levels of IFN-α and TNF-α at any time point post-pretreatment, suggesting that porcine FcγRIIB signal can enhance the innate antiviral response of host cells. PRRSV infection assay mediated by FcγRIIB indicated that selective activation of porcine FcγRIIB in PAM cells enhanced mRNA levels of antiviral cytokine (IFN-α and TNF-α) and repressed mRNA levels of IL-10 in response to PRRSV infection, suggesting that FcγRIIB ligation can enhance the antiviral immune response to PRRSV infection. In addition, FcγRIIB ligation to infection indicated that PRRSV replication in PAM was not positive correlation with increasing of IFN-α mRNA levels and decreasing of IL-10 mRNA levels, suggesting that there is complex viral replication mechanism in immune cells such as PAM for PRRSV.
Collapse
Affiliation(s)
- Yina Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Patel JR, Heldens JGM, Bakonyi T, Rusvai M. Important mammalian veterinary viral immunodiseases and their control. Vaccine 2012; 30:1767-81. [PMID: 22261411 PMCID: PMC7130670 DOI: 10.1016/j.vaccine.2012.01.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 01/03/2012] [Accepted: 01/05/2012] [Indexed: 11/16/2022]
Abstract
This paper offers an overview of important veterinary viral diseases of mammals stemming from aberrant immune response. Diseases reviewed comprise those due to lentiviruses of equine infectious anaemia, visna/maedi and caprine arthritis encephalitis and feline immunodeficiency. Diseases caused by viruses of feline infectious peritonitis, feline leukaemia, canine distemper and aquatic counterparts, Aleutian disease and malignant catarrhal fever. We also consider prospects of immunoprophylaxis for the diseases and briefly other control measures. It should be realised that the outlook for effective vaccines for many of the diseases is remote. This paper describes the current status of vaccine research and the difficulties encountered during their development.
Collapse
Affiliation(s)
- J R Patel
- Jas Biologicals Ltd, 12 Pembroke Avenue, Denny Industrial Estate, Waterbeach, Cambridge CB25 9QR, UK.
| | | | | | | |
Collapse
|
32
|
Qiao S, Jiang Z, Tian X, Wang R, Xing G, Wan B, Bao D, Liu Y, Hao H, Guo J, Zhang G. Porcine FcγRIIb mediates enhancement of porcine reproductive and respiratory syndrome virus (PRRSV) infection. PLoS One 2011; 6:e28721. [PMID: 22220194 PMCID: PMC3248417 DOI: 10.1371/journal.pone.0028721] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 11/14/2011] [Indexed: 01/04/2023] Open
Abstract
Antibody-dependent enhancement (ADE) of virus infection caused by the uptake of virus-antibody complexes by FcγRs is a significant obstacle to the development of effective vaccines to control certain human and animal viral diseases. The activation FcγRs, including FcγRI and FcγRIIa have been shown to mediate ADE infection of virus. In the present paper, we showed that pocine FcγRIIb, an inhibitory FcγR, mediates ADE of PRRSV infection. Stable Marc-145 cell lines expressing poFcγRIIb (Marc-poFcγRII) were established. The relative yield of progeny virus was significantly increased in the presence of sub-neutralization anti-PRRSV antibody. The Fab fragment and normal porcine sera had no effect. Anti-poFcγRII antibody inhibited the enhancement of infection when cells were infected in the presence of anti-PRRSV antibody, but not when cells were infected in the absence of antibody. These results indicate that enhancement of infection in these cells by anti-PRRSV virus antibody is FcγRII-mediated. Identification of the inhibitory FcγR mediating ADE infection should expand our understanding of the mechanisms of pathogenesis for a broad range of infectious diseases and may open many approaches for improvements to the treatment and prevention of such diseases.
Collapse
Affiliation(s)
- Songlin Qiao
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Zhizheng Jiang
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Xiaohui Tian
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Rui Wang
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Guangxu Xing
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Bo Wan
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Dengke Bao
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Yonghui Liu
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Huifang Hao
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Junqing Guo
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - Gaiping Zhang
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, China
- * E-mail:
| |
Collapse
|
33
|
Abstract
The granuloma that forms in response to Mycobacterium tuberculosis must be carefully balanced in terms of immune responses to provide sufficient immune cell activation to inhibit the growth of the bacilli, yet modulate the inflammation to prevent pathology. There are likely many scenarios by which this balance can be reached, given the complexity of the immune responses induced by M. tuberculosis. In this review, we focus on the key role of the macrophage in balancing inflammation in the granuloma.
Collapse
|
34
|
Identification and characterization of a ross river virus variant that grows persistently in macrophages, shows altered disease kinetics in a mouse model, and exhibits resistance to type I interferon. J Virol 2011; 85:5651-63. [PMID: 21430046 DOI: 10.1128/jvi.01189-10] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Alphaviruses, such as chikungunya virus, o'nyong-nyong virus, and Ross River virus (RRV), cause outbreaks of human rheumatic disease worldwide. RRV is a positive-sense single-stranded RNA virus endemic to Australia and Papua New Guinea. In this study, we sought to establish an in vitro model of RRV evolution in response to cellular antiviral defense mechanisms. RRV was able to establish persistent infection in activated macrophages, and a small-plaque variant (RRV(PERS)) was isolated after several weeks of culture. Nucleotide sequence analysis of RRV(PERS) found several nucleotide differences in the nonstructural protein (nsP) region of the RRV(PERS) genome. A point mutation was also detected in the E2 gene. Compared to the parent virus (RRV-T48), RRV(PERS) showed significantly enhanced resistance to beta interferon (IFN-β)-stimulated antiviral activity. RRV(PERS) infection of RAW 264.7 macrophages induced lower levels of IFN-β expression and production than infection with RRV-T48. RRV(PERS) was also able to inhibit type I IFN signaling. Mice infected with RRV(PERS) exhibited significantly enhanced disease severity and mortality compared to mice infected with RRV-T48. These results provide strong evidence that the cellular antiviral response can direct selective pressure for viral sequence evolution that impacts on virus fitness and sensitivity to alpha/beta IFN (IFN-α/β).
Collapse
|
35
|
Kou Z, Lim JY, Beltramello M, Quinn M, Chen H, Liu SN, Martnez-Sobrido L, Diamond MS, Schlesinger JJ, de Silva A, Sallusto F, Jin X. Human antibodies against dengue enhance dengue viral infectivity without suppressing type I interferon secretion in primary human monocytes. Virology 2011; 410:240-7. [DOI: 10.1016/j.virol.2010.11.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 10/04/2010] [Accepted: 11/06/2010] [Indexed: 11/17/2022]
|
36
|
Cell type specificity and host genetic polymorphisms influence antibody-dependent enhancement of dengue virus infection. J Virol 2010; 85:1671-83. [PMID: 21123382 DOI: 10.1128/jvi.00220-10] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Antibody-dependent enhancement (ADE) is implicated in severe, usually secondary, dengue virus (DV) infections. Preexisting heterotypic antibodies, via their Fc-gamma receptor (FcγR) interactions, may increase disease severity through enhanced target cell infection. Greater numbers of infected target cells may contribute to higher viremia and excess cytokine levels often observed in severe disease. Monocytes, macrophages, and immature and mature dendritic cells (DC) are considered major cellular targets of DV. Apheresis of multiple donors allowed isolation of autologous primary myeloid target cell types for head-to-head comparison of infection rates, viral output, and cytokine production under direct infection (without antibody) or ADE conditions (with antibody). All studied cell types except immature DC supported ADE. All cells undergoing ADE secreted proinflammatory cytokines (interleukin-6 [IL-6] and tumor necrosis factor alpha [TNF-α]) at enhancement titers, but distinct cell-type-specific patterns were observed for other relevant proteins (alpha/beta interferon [IFN-α/β] and IL-10). Macrophages produced type I interferons (IFN-α/β) that were modulated by ADE. Mature DC mainly secreted IFN-β. Interestingly, only monocytes secreted IL-10, and only upon antibody-enhanced infection. While ADE infection rates were remarkably consistent in monocytes (10 to 15%) across donors, IL-10 protein levels varied according to previously described regulatory single nucleotide polymorphisms (SNPs) in the IL-10 promoter region. The homozygous GCC haplotype was associated with high-level IL-10 secretion, while the ACC and ATA haplotypes produced intermediate and low levels of IL-10, respectively. Our data suggest that ADE effects are cell type specific, are influenced by host genetics, and, depending on relative infection rates, may further contribute to the complexity of DV pathogenesis.
Collapse
|
37
|
Halstead SB, Mahalingam S, Marovich MA, Ubol S, Mosser DM. Intrinsic antibody-dependent enhancement of microbial infection in macrophages: disease regulation by immune complexes. THE LANCET. INFECTIOUS DISEASES 2010; 10:712-22. [PMID: 20883967 PMCID: PMC3057165 DOI: 10.1016/s1473-3099(10)70166-3] [Citation(s) in RCA: 298] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A wide range of microorganisms can replicate in macrophages, and cell entry of these pathogens via non-neutralising IgG antibody complexes can result in increased intracellular infection through idiosyncratic Fcγ-receptor signalling. The activation of Fcγ receptors usually leads to phagocytosis. Paradoxically, the ligation of monocyte or macrophage Fcγ receptors by IgG immune complexes, rather than aiding host defences, can suppress innate immunity, increase production of interleukin 10, and bias T-helper-1 (Th1) responses to Th2 responses, leading to increased infectious output by infected cells. This intrinsic antibody-dependent enhancement (ADE) of infection modulates the severity of diseases as disparate as dengue haemorrhagic fever and leishmaniasis. Intrinsic ADE is distinct from extrinsic ADE, whereby complexes of infectious agents with non-neutralising antibodies lead to an increased number of infected cells. Intrinsic ADE might be involved in many protozoan, bacterial, and viral infections. We review insights into intracellular mechanisms and implications of enhanced pathogenesis after ligation of macrophage Fcγ receptors by infectious immune complexes.
Collapse
|
38
|
Enterovirus 71 infection of monocytes with antibody-dependent enhancement. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1517-23. [PMID: 20685937 DOI: 10.1128/cvi.00108-10] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Enterovirus (EV) is an RNA virus that has circulated with different serotypes and genotypes worldwide. Enterovirus 71 (EV71) is a major neurotropic virus that causes severe brain stem encephalitis (BE) in infants and young children. The most vulnerable age for fatal infection is 6 to 11 months. This is associated with the coincident decline in maternal antibodies. The current report describes our finding that EV71 can infect human peripheral blood monocytes. We were able to show that EV71 infection is enhanced in the monocytic cell line THP-1 by the presence of subneutralizing concentrations of anti-EV71 antibodies. We also found that antibody-dependent enhancement (ADE) is mediated in part by Fcγ receptors. These observations support the concept that ADE augments the infectivity of EV71 for human monocytes and contributes to the age-dependent pathogenesis of EV71-induced disease. The ADE phenomenon must be considered during the development of an EV71 vaccine.
Collapse
|
39
|
Huang Y, Meng X. Novel strategies and approaches to develop the next generation of vaccines against porcine reproductive and respiratory syndrome virus (PRRSV). Virus Res 2010; 154:141-9. [PMID: 20655962 PMCID: PMC7132426 DOI: 10.1016/j.virusres.2010.07.020] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 07/15/2010] [Indexed: 12/15/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is an economically important swine pathogen. Since its discovery in the early 1990s, tremendous progresses have been made in understanding the molecular biology and pathogenesis of PRRSV. Although modified live-attenuated vaccines (MLVs) and inactivated vaccines against PRRSV have been available for more than a decade, the disease remains difficult to control. The efficacies of these vaccines especially against heterologous strains remain questionable: the MLVs were generally effective against homologous strains but variable in success against heterologous strains, and the outcomes of inactivated vaccines in the field are not very promising. With the development of PRRSV reverse genetics systems and the acquisition of new understanding on anti-PRRSV immunity, rational design of the next generation of PRRSV vaccines can now be explored. In this review, we discussed the recent advances in anti-PRRSV immunity and vaccinology, the recent progresses in PRRSV vaccine development particularly the reverse genetics system-based vaccine development, and provided a perspective on potential novel strategies and approaches that may be applicable to the development of the next generation of PRRSV vaccines.
Collapse
Affiliation(s)
| | - X.J. Meng
- Corresponding author at: Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Polytechnic Institute and State University, 1981 Kraft Drive, Room 2036, Blacksburg, VA 24061-0913, USA. Tel.: +1 540 231 6912; fax: +1 540 231 3426.
| |
Collapse
|
40
|
Abstract
Chikungunya virus is a mosquito-borne arthrogenic alphavirus that has recently reemerged to produce the largest epidemic ever documented for this virus. Here we describe a new adult wild-type mouse model of chikungunya virus arthritis, which recapitulates the self-limiting arthritis, tenosynovitis, and myositis seen in humans. Rheumatic disease was associated with a prolific infiltrate of monocytes, macrophages, and NK cells and the production of monocyte chemoattractant protein 1 (MCP-1), tumor necrosis factor alpha (TNF-alpha), and gamma interferon (IFN-gamma). Infection with a virus isolate from the recent Reunion Island epidemic induced significantly more mononuclear infiltrates, proinflammatory mediators, and foot swelling than did an Asian isolate from the 1960s. Primary mouse macrophages were shown to be productively infected with chikungunya virus; however, the depletion of macrophages ameliorated rheumatic disease and prolonged the viremia. Only 1 microg of an unadjuvanted, inactivated, whole-virus vaccine derived from the Asian isolate completely protected against viremia and arthritis induced by the Reunion Island isolate, illustrating that protection is not strain specific and that low levels of immunity are sufficient to mediate protection. IFN-alpha treatment was able to prevent arthritis only if given before infection, suggesting that IFN-alpha is not a viable therapy. Prior infection with Ross River virus, a related arthrogenic alphavirus, and anti-Ross River virus antibodies protected mice against chikungunya virus disease, suggesting that individuals previously exposed to Ross River virus should be protected from chikungunya virus disease. This new mouse model of chikungunya virus disease thus provides insights into pathogenesis and a simple and convenient system to test potential new interventions.
Collapse
|
41
|
Abstract
Much remains to be learned about the pathogenesis of the different manifestations of dengue virus (DENV) infections in humans. They may range from subclinical infection to dengue fever, dengue hemorrhagic fever (DHF), and eventually dengue shock syndrome (DSS). As both cell tropism and tissue tropism of DENV are considered major determinants in the pathogenesis of dengue, there is a critical need for adequate tropism assays, animal models, and human autopsy data. More than 50 years of research on dengue has resulted in a host of literature, which strongly suggests that the pathogenesis of DHF and DSS involves viral virulence factors and detrimental host responses, collectively resulting in abnormal hemostasis and increased vascular permeability. Differential targeting of specific vascular beds is likely to trigger the localized vascular hyperpermeability underlying DSS. A personalized approach to the study of pathogenesis will elucidate the basis of individual risk for development of DHF and DSS as well as identify the genetic and environmental bases for differences in risk for development of severe disease.
Collapse
|
42
|
Abstract
A large range of human viruses are associated with the development of arthritis or arthralgia. Although there are many parallels with autoimmune arthritides, there is little evidence that viral arthritides lead to autoimmune disease. In humans viral arthritides usually last from weeks to months, can be debilitating, and are usually treated with non-steroidal anti-inflammatory drugs, but with variable success. Viral arthritides likely arise from immunopathological inflammatory responses directed at viruses and/or their products residing and/or replicating within joint tissues. Macrophages recruited by monocyte chemoattractant protein-1 (MCP-1/CCL2) and activated by interferon, and proinflammatory mediators like tumour necrosis factor alpha, interferon gamma, interleukin-6 and interleukin-1beta appear to be common elements in this group of diseases. The challenge for new treatments is to target excessive inflammation without compromising anti-viral immunity. Recent evidence from mouse models suggests targeting MCP-1 or complement may emerge as viable new treatment options for viral arthritides.
Collapse
|
43
|
|
44
|
Foong YY, Jans DA, Rolph MS, Gahan ME, Mahalingam S. Interleukin-15 mediates potent antiviral responses via an interferon-dependent mechanism. Virology 2009; 393:228-37. [PMID: 19729181 DOI: 10.1016/j.virol.2009.07.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2009] [Revised: 04/20/2009] [Accepted: 07/06/2009] [Indexed: 10/20/2022]
Abstract
Interleukin-15 (IL-15) is a potent growth factor for activated T and natural killer (NK) cells, stimulator of memory T cells and plays an important role in viral immunity. To investigate mechanisms underlying the antiviral activity of IL-15, a recombinant vaccinia virus (rVV) encoding murine IL-15 (VV-IL-15) was constructed. Following infection of mice with VV-IL-15, virus titres in the ovaries were significantly reduced compared to mice infected with control VV. Growth of VV-IL-15 was also reduced in nude athymic mice, indicating the antiviral activity of IL-15 does not require T cells. Additionally, VV-IL-15 augmented the cytolytic activity of natural NK cells in the spleen and enhanced interferon (IFN) mRNA expression and transcription factors associated with IFN induction. Using knockout mice and antibody depletion studies, we showed for the first time that the control of VV-IL-15 replication in mice is dependent on NK cells and IFNs and, in their absence, the protective role of IL-15 is abolished.
Collapse
Affiliation(s)
- Y Y Foong
- Division of Immunology and Genetics, The John Curtin School of Medical Research, Australian National University, Canberra ACT 0200, Australia
| | | | | | | | | |
Collapse
|
45
|
Long HT, Hibberd ML, Hien TT, Dung NM, Van Ngoc T, Farrar J, Wills B, Simmons CP. Patterns of gene transcript abundance in the blood of children with severe or uncomplicated dengue highlight differences in disease evolution and host response to dengue virus infection. J Infect Dis 2009; 199:537-546. [PMID: 19138155 DOI: 10.1086/596507] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
DNA microarrays and specific reverse-transcription polymerase chain reaction assays were used to reveal transcriptional patterns in the blood of children presenting with dengue shock syndrome (DSS) and well-matched patients with uncomplicated dengue. The transcriptome of patients with acute uncomplicated dengue was characterized by a metabolically demanding "host-defense" profile; transcripts related to oxidative metabolism, interferon signaling, protein ubiquination, apoptosis, and cytokines were prominent. In contrast, the transcriptome of patients with DSS was surprisingly benign, particularly with regard to transcripts derived from apoptotic and type I interferon pathways. These data highlight significant heterogeneity in the type or timing of host transcriptional immune responses precipitated by dengue virus infection independent of the duration of illness. In particular, they suggest that, if transcriptional events in the blood compartment contribute to capillary leakage leading to hypovolemic shock, they occur before cardiovascular decompensation, a finding that has implications for rational adjuvant therapy in this syndrome.
Collapse
Affiliation(s)
| | | | - Tran Tinh Hien
- Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | | | - Tran Van Ngoc
- Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Jeremy Farrar
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam.,Centre for Tropical Medicine, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Bridget Wills
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam.,Centre for Tropical Medicine, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| | - Cameron P Simmons
- Oxford University Clinical Research Unit, Ho Chi Minh City, Vietnam.,Centre for Tropical Medicine, University of Oxford, Churchill Hospital, Oxford, United Kingdom
| |
Collapse
|
46
|
Rodrigo WWSI, Rodrigo WWIS, Alcena DC, Kou Z, Kochel TJ, Porter KR, Comach G, Rose RC, Jin X, Schlesinger JJ. Difference between the abilities of human Fcgamma receptor-expressing CV-1 cells to neutralize American and Asian genotypes of dengue virus 2. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 16:285-7. [PMID: 19038781 PMCID: PMC2643537 DOI: 10.1128/cvi.00363-08] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 11/04/2008] [Accepted: 11/17/2008] [Indexed: 01/08/2023]
Abstract
Sera from patients involved in a Peruvian outbreak of dengue virus serotype 1 infection cross-neutralized the American genotype of dengue virus serotype 2 up to 100-fold more efficiently than they did the virulent Asian genotype of dengue virus serotype 2, as determined by a plaque reduction neutralization test (PRNT) with CV-1 fibroblasts modified to express human Fcgamma receptor CD32. The concordant preferential immune enhancement of the Asian genotype of dengue virus serotype 2 in human monocytes suggests that such a modification might strengthen the correlation between the PRNT titer and protection.
Collapse
Affiliation(s)
- W W Shanaka I Rodrigo
- Department of Pathology, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Macrophages display remarkable plasticity and can change their physiology in response to environmental cues. These changes can give rise to different populations of cells with distinct functions. In this Review we suggest a new grouping of macrophage populations based on three different homeostatic activities - host defence, wound healing and immune regulation. We propose that similarly to primary colours, these three basic macrophage populations can blend into various other 'shades' of activation. We characterize each population and provide examples of macrophages from specific disease states that have the characteristics of one or more of these populations.
Collapse
Affiliation(s)
- David M Mosser
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland 20742, USA.
| | | |
Collapse
|
48
|
Huisman W, Martina BEE, Rimmelzwaan GF, Gruters RA, Osterhaus ADME. Vaccine-induced enhancement of viral infections. Vaccine 2008; 27:505-12. [PMID: 19022319 PMCID: PMC7131326 DOI: 10.1016/j.vaccine.2008.10.087] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2008] [Revised: 10/29/2008] [Accepted: 10/29/2008] [Indexed: 12/19/2022]
Abstract
Examples of vaccine-induced enhancement of susceptibility to virus infection or of aberrant viral pathogenesis have been documented for infections by members of different virus families. Several mechanisms, many of which still are poorly understood, are at the basis of this phenomenon. Vaccine development for lentivirus infections in general, and for HIV/AIDS in particular, has been little successful. Certain experimental lentiviral vaccines even proved to be counterproductive: they rendered vaccinated subjects more susceptible to infection rather than protecting them. For vaccine-induced enhanced susceptibility to infection with certain viruses like feline coronavirus, Dengue virus, and feline immunodeficiency virus, it has been shown that antibody-dependent enhancement (ADE) plays an important role. Other mechanisms may, either in the absence of or in combination with ADE, be involved. Consequently, vaccine-induced enhancement has been a major stumble block in the development of certain flavi-, corona-, paramyxo-, and lentivirus vaccines. Also recent failures in the development of a vaccine against HIV may at least in part be attributed to induction of enhanced susceptibility to infection. There may well be a delicate balance between the induction of protective immunity on the one hand and the induction of enhanced susceptibility on the other. The present paper reviews the currently known mechanisms of vaccine-induced enhancement of susceptibility to virus infection or of aberrant viral pathogenesis.
Collapse
Affiliation(s)
- W Huisman
- Erasmus MC, Institute of Virology, P.O. Box 1738, 3000 DR Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
49
|
Lidbury BA, Rulli NE, Suhrbier A, Smith PN, McColl SR, Cunningham AL, Tarkowski A, van Rooijen N, Fraser RJ, Mahalingam S. Macrophage-derived proinflammatory factors contribute to the development of arthritis and myositis after infection with an arthrogenic alphavirus. J Infect Dis 2008; 197:1585-93. [PMID: 18433328 DOI: 10.1086/587841] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Alphaviruses, such as chikungunya virus and Ross River virus (RRV), are associated with outbreaks of infectious rheumatic disease in humans worldwide. Using an established mouse model of disease that mimics RRV disease in humans, we showed that macrophage-derived factors are critical in the development of striated muscle and joint tissue damage. Histologic analyses of muscle and ankle joint tissues demonstrated a substantial reduction in inflammatory infiltrates in infected mice depleted of macrophages (i.e., "macrophage-depleted mice"). Levels of the proinflammatory factors tumor necrosis factor-alpha, interferon-gamma, and macrophage chemoattractant protein-1 were also dramatically reduced in tissue samples obtained from infected macrophage-depleted mice, compared with samples obtained from infected mice without macrophage depletion. These factors were also detected in the synovial fluid of patients with RRV-induced polyarthritis. Neutralization of these factors reduced the severity of disease in mice, whereas blocking nuclear factor kappaB by treatment with sulfasalazine ameliorated RRV inflammatory disease and tissue damage. To our knowledge, these findings are the first to demonstrate that macrophage-derived products play important roles in the development of arthritis and myositis triggered by alphavirus infection.
Collapse
Affiliation(s)
- Brett A Lidbury
- Virus and Inflammation Research Group, Faculty of Sciences, University of Canberra, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Jin X. Cellular and molecular basis of antibody-dependent enhancement in human dengue pathogenesis. Future Virol 2008. [DOI: 10.2217/17460794.3.4.343] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Dengue fever is gaining increased attention as a major global health problem. It occurs annually in 50–100 million people in more than 100 countries, and places half a million people at risk of life-threatening diseases: dengue hemorrhagic fever and dengue shock syndrome (DHF/DSS). The pathogenic mechanisms causing DHF/DSS are not clearly understood. This article reviews cellular and molecular mechanisms that might be responsible for the initiation of the pathogenic processes, including hypotheses for DHF/DSS, dengue-permissive target cells, putative dengue receptors, neutralizing and enhancing antibodies to dengue virus, mechanisms of vascular plasma leakage, innate immune response in dengue infection and antibody-dependent enhancement of dengue infection. While reviewing the literature, the article also gives the author’s opinion on perceived areas of importance for future research in human dengue pathogenesis.
Collapse
Affiliation(s)
- Xia Jin
- Department of Medicine, Infectious Diseases Division, University of Rochester Medical Center, 601 Elmwood Avenue, Box 689, Room 3-5103, Rochester, NY 14642, USA
| |
Collapse
|