1
|
Chopra A, Venugopalan A. Chikungunya and other viral arthritis. Best Pract Res Clin Rheumatol 2025:102068. [PMID: 40360316 DOI: 10.1016/j.berh.2025.102068] [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: 02/18/2025] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025]
Abstract
Several viruses cause acute and chronic arthritis. Millions of people suffered from Chikungunya(CHIK) during the recent epidemics/outbreaks in Asia, Africa and the Americas. Almost 20-40 % failed to recover completely and suffered from chronic pain and arthritis sequel. A wide spectrum of clinical phenotypic arthritis was described. Non-specific arthralgias(NSA) and soft tissue pains were predominant although inflammatory arthritis (mostly undifferentiated)(IA-U) was substantial. Specifically, rheumatoid arthritis(RA) and spondyloarthritis(SpA) like disorders were described. The frequency of biomarkers such as rheumatoid factor(RF) was low. Arthritis was mostly non-erosive in population studies. Abnormal immune mechanisms and persistent specific CHIK virus (CHIKV) IgM and IgG antibodies were shown. The etiopathogenetic evidence was divided between intense joint tissue inflammation due to prolonged virus persistence and abnormal autoimmune mechanisms. There was no specific therapy. The symptomatic management was often combined with an empirical use of disease modifying anti rheumatoid drugs and steroids. Substantial research is required to address knowledge gaps and unravel evidence-based medicine.
Collapse
Affiliation(s)
- Arvind Chopra
- Center for Rheumatic Diseases, 11 Hermes Elegance, 1988 Convent Street, Camp, Pune, 411001, India.
| | - Anuradha Venugopalan
- Center for Rheumatic Diseases, 11 Hermes Elegance, 1988 Convent Street, Camp, Pune, 411001, India.
| |
Collapse
|
2
|
de Souza WM, Lecuit M, Weaver SC. Chikungunya virus and other emerging arthritogenic alphaviruses. Nat Rev Microbiol 2025:10.1038/s41579-025-01177-8. [PMID: 40335675 DOI: 10.1038/s41579-025-01177-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2025] [Indexed: 05/09/2025]
Abstract
Arthritogenic alphaviruses are arboviruses (arthropod-borne viruses) that are genetically and serologically related positive-strand RNA viruses and cause epidemics on a global scale. They are transmitted by mosquitoes and cause diseases in humans that are mainly characterized by fever and often debilitating, sometimes chronic polyarthralgia. At present, approved treatments or vaccines are not available for most arthritogenic alphaviruses, and recently licensed vaccines against chikungunya virus are awaiting implementation in endemic areas. Most arthritogenic alphaviruses are currently limited to specific geographic areas due to vector distributions and availability of amplifying hosts, but they pose a substantial risk of emergence in other regions. The exception is chikungunya virus, which has emerged repeatedly from Africa, established sustained and efficient transmission in urban areas (including in temperate climates) and has caused major epidemics across the world. In this Review, we highlight recent advances in our understanding of the transmission cycles of arthritogenic alphaviruses, their vectors, epidemiology, transmission dynamics, evolution, pathophysiology and immune responses. We also outline strategies and countermeasures to anticipate and mitigate the impact of arthritogenic alphaviruses on human health.
Collapse
Affiliation(s)
- William M de Souza
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, KY, USA
| | - Marc Lecuit
- Institut Pasteur, Université Paris Cité, Inserm U1117, Biology of Infection Unit, Paris, France
- Department of Infectious Diseases and Tropical Medicine, Assistance Publique-Hôpitaux de Paris, Institut Imagine, Necker-Enfants Malades University Hospital, Paris, France
| | - Scott C Weaver
- World Reference Center for Emerging Viruses and Arboviruses, Institute for Human Infections and Immunity, and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
3
|
Gonçalves WA, de Sousa CDF, Teixeira MM, Souza DG. A brief overview of chikungunya-related pain. Eur J Pharmacol 2025; 994:177322. [PMID: 39892450 DOI: 10.1016/j.ejphar.2025.177322] [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: 07/28/2024] [Revised: 01/15/2025] [Accepted: 01/29/2025] [Indexed: 02/03/2025]
Abstract
Pain is an important symptom associated with the arboviral disease caused by the Chikungunya virus (CHIKV). For a significant number of patients, this symptom can persist for months or even years, negatively affecting their quality of life. Unfortunately, pharmacological options for this condition are limited and only partially effective, as the underlying mechanisms associated with CHIKV-induced pain are still poorly understood. The re-emergence of CHIKV has led to new outbreaks, and the expected high prevalence of pain in these global events requires new scientific advances to find more effective solutions. Here we review the main aspects of pain caused by CHIKV infection, such as the anatomy of the affected sites, the prevalence and management of this symptom, the diversity of possible cellular and molecular mechanisms, and finally highlight a promising meningeal pathway to elucidate the mechanisms involved in the unsolved problem of CHIKV-associated pain.
Collapse
Affiliation(s)
- William Antonio Gonçalves
- Departamento de Morfologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| | - Carla Daiane Ferreira de Sousa
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, University of Münster, Münster, Germany.
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| | - Daniele G Souza
- Laboratório Interação Microrganismo Hospedeiro, Departamento de Microbiologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.
| |
Collapse
|
4
|
Salomão NG, Araújo L, de Souza LJ, Luiza Young A, Basílio-de-Oliveira C, Basílio-de-Oliveira RP, de Carvalho JJ, Nunes PCG, da Silva Amorim JF, Barbosa DVDS, Paes MV, Rabelo K, Dos Santos F. Chikungunya virus infection in the skin: histopathology and cutaneous immunological response. Front Microbiol 2025; 16:1497354. [PMID: 39935638 PMCID: PMC11811090 DOI: 10.3389/fmicb.2025.1497354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
Alphavirus chikungunya virus (CHIKV) is an arbovirus, belonging to the Togaviridae family. The disease caused by CHIKV generally evolves with spontaneous resolution in a few weeks; however, progression to a chronic disease may occur. The most common symptoms are fever, myalgia, and arthralgia; however, skin manifestations may occur in 40 to 80% of infected individuals. Morbilliform and maculopapular erythematous eruptions, vesiculobullous lesions, generalized erythema, maculopapular eruption and skin peeling, hypermelanosis, painful oral lesions, and urticarial lesions have been reported. Usually, these manifestations disappear, but they can become sequelae. Since the skin is the first line of defense against CHIKV infection, in this study, we aimed to investigate the immunohistopathological aspects of the skin of infected individuals during the acute phase of the disease by performing histopathological and ultrastructural analysis, detection and quantification of the viral genome, detection of viral antigen and immune cells, and cytokines/chemokines' characterization. The main histopathological findings were perivascular and inflammatory infiltrates, blood capillary ectasia, and interstitial edema. The immunohistochemistry revealed CHIKV antigen in the epidermis, endothelial cells, fibroblasts, and macrophages in the reticular and papillary dermis; inflammatory cells infiltrate; arrector pili muscle; sweat and sebaceous glands; and hair follicle. Moreover, inflammatory infiltrates were composed of lymphocytes (CD4+ and CD8+) and macrophages (CD68+) in the dermis and perivascular infiltrate. TNF-α, IL-6, RANTES, and VEGFR2 were expressed in the epidermis, blood vessels, sweat glands, and migrating cells. Loss of contact among adjacent keratinocytes, epidermis presenting necrotic cells, and fibroblasts with dilated cisternae in the endoplasmic reticulum and mitochondria with few cristae was observed by transmission electron microscopy. Studies involving skin immunopathogenesis during CHIKV infection are still scarce; therefore, the findings presented here can contribute to a better understanding of the disease immunopathogenesis.
Collapse
Affiliation(s)
- Natália Gedeão Salomão
- Laboratório das Interações Vírus-Hospedeiros, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, Brazil
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, Brazil
| | - Luciana Araújo
- Departamento de Anatomia Patológica, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | | | | | - Carlos Basílio-de-Oliveira
- Departamento de Anatomia Patológica, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | | | - Jorge José de Carvalho
- Laboratório de Ultraestrutura e Biologia Tecidual, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | - Priscilla Conrado Guerra Nunes
- Laboratório das Interações Vírus-Hospedeiros, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, Brazil
| | | | | | - Marciano Viana Paes
- Laboratório das Interações Vírus-Hospedeiros, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, Brazil
| | - Kíssila Rabelo
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, Brazil
- Laboratório de Ultraestrutura e Biologia Tecidual, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brazil
| | - Flavia Dos Santos
- Laboratório das Interações Vírus-Hospedeiros, Instituto Oswaldo Cruz/Fundação Oswaldo Cruz (IOC/Fiocruz), Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Paine DN, Hermance M, Thangamani S. Early transcriptomic changes at the skin interface during Powassan virus transmission by Ixodes scapularis ticks. Front Immunol 2025; 15:1511132. [PMID: 39872517 PMCID: PMC11769802 DOI: 10.3389/fimmu.2024.1511132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 12/10/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction Powassan virus (POWV), a vector-borne pathogen transmitted by Ixodes ticks in North America, is the causative agent of Powassan encephalitis. As obligate hematophagous organisms, ticks transmit pathogens like POWV at the tick bite site, specifically during the initial stages of feeding. Tick feeding and salivary factors modulate the host's immunological responses, facilitating blood feeding and pathogen transmission. However, the mechanisms of immunomodulation during POWV transmission remain inadequately understood. In this study, we investigated the global cutaneous transcriptomic changes associated with tick bites during POWV transmission. Methods We collected skin biopsies from the tick attachment sites at 1, 3, and 6 h after feeding by POWV-infected and uninfected ticks, followed by RNA sequencing of these samples. Differentially expressed genes were analyzed for pathway enrichment using gene ontology and pathway enrichment analyses. Results Our findings reveal that tick feeding alone significantly impacts the skin transcriptome within the first 1 to 3 h of tick attachment. Although early POWV transmission induces minimal changes in the local environment, a pronounced shift toward a proinflammatory state is observed 6 h after tick attachment, characterized by neutrophil recruitment and interleukin signaling. Discussion These transcriptomic data elucidate the dynamic changes at the tick bite site, transitioning from changes that assist blood meal acquisition to a proinflammatory phase that may facilitate viral dissemination.
Collapse
Affiliation(s)
- Dakota N. Paine
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, United States
- State University of New York Center for Vector-Borne Diseases, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Meghan Hermance
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL, United States
| | - Saravanan Thangamani
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, NY, United States
- State University of New York Center for Vector-Borne Diseases, State University of New York Upstate Medical University, Syracuse, NY, United States
- Upstate Global Health Institute, State University of New York Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
6
|
Lozano-Parra A, Herrera V, Urcuqui-Inchima S, Ramírez RMG, Villar LÁ. Acute Immunological Profile and Prognostic Biomarkers of Persistent Joint Pain in Chikungunya Fever: A Systematic Review. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2024; 97:473-489. [PMID: 39703607 PMCID: PMC11650912 DOI: 10.59249/rqyj3197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Chikungunya virus infection (CHIKV) increases the risk of persistent arthralgia; however, there is no consistent evidence regarding prognostic biomarkers of progression to chronic arthropathy. This systematic review provides an overview of currently available literature about the potential role of the acute immunologic response in predicting long-term joint pain in patients with a diagnosis of CHIKV. We searched for observational studies using the terms "chikungunya," "cytokines," "biomarkers," and "joint pain" in PubMed/MEDLINE, LILACS, Cochrane Library Plus, and SCOPUS databases, restricting to articles published in English and up to April 2024. PROSPERO registration number: CRD42021279400. Thirty-eight studies were selected for qualitative synthesis with a maximum duration from diagnosis to clinical evaluation of 60 months. The sample sizes ranged from 8 to 346 participants (age range: 0-90 years). We identified an immunologic profile during the acute phase of CHIKV that includes increased levels of proinflammatory cytokines (IFN-α, IFN-γ, IL-2R, IL-6, IL-7, and IL-8), anti-inflammatory cytokines (IL-1Ra and IL-4), chemokines (MCP-1, MIG, and IP-10) and growth factors (VEGF and G-CSF). Only one out of two studies reported differences in cytokine levels during the acute phase, predicting persistent joint pain at 20 months of follow-up. Also, persistence of anti-CHIKV IgG seemed to be a potential prognostic marker. The evidence suggests the existence of an inflammatory response in the acute phase of CHIKV that persists during its chronic phase; however, there is no unequivocal candidate set of biomarkers of progression toward long-term articular sequelae.
Collapse
Affiliation(s)
- Anyela Lozano-Parra
- Grupo Epidemiología Clínica, Escuela de Medicina,
Universidad Industrial de Santander UIS, Bucaramanga, Colombia
| | - Víctor Herrera
- Grupo Epidemiología Clínica, Escuela de Medicina,
Universidad Industrial de Santander UIS, Bucaramanga, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Epidemiología Clínica, Escuela de Medicina,
Universidad Industrial de Santander UIS, Bucaramanga, Colombia
- Grupo Inmunovirología, Departamento de Microbiología y
Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín,
Colombia
| | | | - Luis Ángel Villar
- Centro de Atención y Diagnóstico de Enfermedades
Infecciosas (CDI), Fundación INFOVIDA, Bucaramanga, Colombia
| |
Collapse
|
7
|
Landers VD, Thomas M, Isom CM, Karki D, Sokoloski KJ. Capsid protein mediated evasion of IRAK1-dependent signalling is essential to Sindbis virus neuroinvasion and virulence in mice. Emerg Microbes Infect 2024; 13:2300452. [PMID: 38164715 PMCID: PMC10773654 DOI: 10.1080/22221751.2023.2300452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
ABSTRACTAlphaviruses are arthropod-borne, single-stranded positive-sense RNA viruses that are recognized as rapidly emerging pathogens. Despite being exquisitely sensitive to the effects of the innate immune response alphaviruses can readily replicate, disseminate, and induce pathogenesis in immunologically competent hosts. Nonetheless, how alphaviruses evade the induction of an innate immune response prior to viral gene expression, or in non-permissive infections, is unknown. Previously we reported the identification of a novel host/pathogen interaction between the viral Capsid (CP) protein and the host IRAK1 protein. The CP/IRAK1 interaction was determined to negatively impact IRAK1-dependent PAMP detection in vitro, however, the precise importance of the CP/IRAK1 interaction to alphaviral infection remained unknown. Here we detail the identification of the CP/IRAK1 interaction determinants of the Sindbis virus (SINV) CP protein and examine the importance of the interaction to alphaviral infection and pathogenesis in vivo using an interaction deficient mutant of the model neurotropic strain of SINV. Importantly, these interaction determinants are highly conserved across multiple Old-World alphaviruses, including Ross River virus (RRV), Mayaro virus (MAYV), Chikungunya virus (CHIKV), and Semliki Forest virus (SFV). In the absence of a functional CP/IRAK1 interaction, SINV replication is significantly restricted and fails to disseminate from the primary site of inoculation due to the induction of a robust type-I Interferon response. Altogether these data indicate that the evasion of IRAK1-dependent signalling is critical to overcoming the host innate immune response and the in vivo data presented here demonstrate the importance of the CP/IRAK1 interaction to neurovirulence and pathogenesis.
Collapse
Affiliation(s)
- V Douglas Landers
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Milton Thomas
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Cierra M. Isom
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Deepa Karki
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Kevin J. Sokoloski
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, KY, USA
- Center for Predictive Medicine and Emerging Infectious Diseases, University of Louisville, Louisville, KY, USA
| |
Collapse
|
8
|
Freppel W, Silva LA, Stapleford KA, Herrero LJ. Pathogenicity and virulence of chikungunya virus. Virulence 2024; 15:2396484. [PMID: 39193780 PMCID: PMC11370967 DOI: 10.1080/21505594.2024.2396484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted, RNA virus that causes an often-severe musculoskeletal illness characterized by fever, joint pain, and a range of debilitating symptoms. The virus has re-emerged as a global health threat in recent decades, spreading from its origin in Africa across Asia and the Americas, leading to widespread outbreaks impacting millions of people. Despite more than 50 years of research into the pathogenesis of CHIKV, there is still no curative treatment available. Current management of CHIKV infections primarily involves providing supportive care to alleviate symptoms and improve the patient's quality of life. Given the ongoing threat of CHIKV, there is an urgent need to better understand its pathogenesis. This understanding is crucial for deciphering the mechanisms underlying the disease and for developing effective strategies for both prevention and management. This review aims to provide a comprehensive overview of CHIKV and its pathogenesis, shedding light on the complex interactions of viral genetics, host factors, immune responses, and vector-related factors. By exploring these intricate connections, the review seeks to contribute to the knowledge base surrounding CHIKV, offering insights that may ultimately lead to more effective prevention and management strategies for this re-emerging global health threat.
Collapse
Affiliation(s)
- Wesley Freppel
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, Australia
| | - Laurie A. Silva
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kenneth A. Stapleford
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Lara J. Herrero
- Institute for Biomedicine and Glycomics, Gold Coast Campus, Griffith University, Southport, Australia
| |
Collapse
|
9
|
Fritsch H, Giovanetti M, Clemente LG, da Rocha Fernandes G, Fonseca V, de Lima MM, Falcão M, de Jesus N, de Cerqueira EM, Venâncio da Cunha R, de Oliveira Francisco MVL, de Siqueira IC, de Oliveira C, Xavier J, Ferreira JGG, Queiroz FR, Smith E, Tisoncik-Go J, Van Voorhis WC, Rabinowitz PM, Wasserheit JN, Gale M, de Filippis AMB, Alcantara LCJ. Unraveling the Complexity of Chikungunya Virus Infection Immunological and Genetic Insights in Acute and Chronic Patients. Genes (Basel) 2024; 15:1365. [PMID: 39596565 PMCID: PMC11593632 DOI: 10.3390/genes15111365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Background: The chikungunya virus (CHIKV), transmitted by infected Aedes mosquitoes, has caused a significant number of infections worldwide. In Brazil, the emergence of the CHIKV-ECSA genotype in 2014 posed a major public health challenge due to its association with more severe symptoms. Objectives/Methods: This study aimed to shed new light on the host immune response by examining the whole-blood transcriptomic profile of both CHIKV-acute and chronically infected individuals from Feira de Santana, Bahia, Brazil, a region heavily affected by CHIKV, Dengue, and Zika virus epidemics. Results: Our data reveal complex symptomatology characterized by arthralgia and post-chikungunya neuropathy in individuals with chronic sequelae, particularly affecting women living in socially vulnerable situations. Analysis of gene modules suggests heightened metabolic processes, represented by an increase in NADH, COX5A, COA3, CYC1, and cap methylation in patients with acute disease. In contrast, individuals with chronic manifestations exhibit a distinct pattern of histone methylation, probably mediated by NCOA3 in the coactivation of different nuclear receptors, KMT2 genes, KDM3B and TET2, and with alterations in the immunological response, majorly led by IL-17RA, IL-6R, and STAT3 Th17 genes. Conclusion: Our results emphasize the complexity of CHIKV disease progression, demonstrating the heterogeneous gene expression and symptomatologic scenario across both acute and chronic phases. Moreover, the identification of specific gene modules associated with viral pathogenesis provides critical insights into the molecular mechanisms underlying these distinct clinical manifestations.
Collapse
Affiliation(s)
- Hegger Fritsch
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (H.F.); (J.X.)
- Institut National de la Santé et de la Recherche Médicale, U1259—MAVIVHe, Université de Tours, 37032 Tours, France
| | - Marta Giovanetti
- Department of Science and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128 Rome, Italy;
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil;
- Climate Amplified Diseases and Epidemics (CLIMADE)—CLIMADE Americas, Belo Horizonte 30190-002, Brazil
| | - Luan Gaspar Clemente
- Escola Superior de Agricultura Luiz de Queiroz, Departamento de Zootecnia, Universidade de São Paulo, Piracicaba 13418-900, Brazil;
| | | | - Vagner Fonseca
- Departamento de Ciências Exatas e da Terra, Universidade Estadual da Bahia, Salvador 41150-000, Brazil;
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Maricelia Maia de Lima
- Departamento de Saúde, Universidade Estadual de Feira de Santana, Feira de Santana 44036-900, Brazil; (M.M.d.L.); (E.M.d.C.)
- Secretaria de Municipal de Saúde de Feira de Santana, Divisão de Vigilância Epidemiológica, Feira de Santana 44027-010, Brazil; (M.F.); (N.d.J.)
| | - Melissa Falcão
- Secretaria de Municipal de Saúde de Feira de Santana, Divisão de Vigilância Epidemiológica, Feira de Santana 44027-010, Brazil; (M.F.); (N.d.J.)
| | - Neuza de Jesus
- Secretaria de Municipal de Saúde de Feira de Santana, Divisão de Vigilância Epidemiológica, Feira de Santana 44027-010, Brazil; (M.F.); (N.d.J.)
| | - Erenilde Marques de Cerqueira
- Departamento de Saúde, Universidade Estadual de Feira de Santana, Feira de Santana 44036-900, Brazil; (M.M.d.L.); (E.M.d.C.)
| | | | | | | | - Carla de Oliveira
- Laboratório de Arbovírus e Vírus Hemorrágicos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil;
| | - Joilson Xavier
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (H.F.); (J.X.)
| | - Jorge Gomes Goulart Ferreira
- Núcleo de Ensino, Pesquisa e Inovação, Instituto Mário Penna, Belo Horizonte 30380-420, Brazil; (J.G.G.F.); (F.R.Q.)
| | - Fábio Ribeiro Queiroz
- Núcleo de Ensino, Pesquisa e Inovação, Instituto Mário Penna, Belo Horizonte 30380-420, Brazil; (J.G.G.F.); (F.R.Q.)
| | - Elise Smith
- Department of Immunology, University of Washington, Seattle, WA 98109, USA; (E.S.); (J.T.-G.); (M.G.J.)
| | - Jennifer Tisoncik-Go
- Department of Immunology, University of Washington, Seattle, WA 98109, USA; (E.S.); (J.T.-G.); (M.G.J.)
| | | | - Peter M. Rabinowitz
- Departments of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA;
| | | | - Michael Gale
- Department of Immunology, University of Washington, Seattle, WA 98109, USA; (E.S.); (J.T.-G.); (M.G.J.)
| | - Ana Maria Bispo de Filippis
- Laboratório de Arbovírus e Vírus Hemorrágicos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil;
| | - Luiz Carlos Junior Alcantara
- Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte 30190-002, Brazil;
- Climate Amplified Diseases and Epidemics (CLIMADE)—CLIMADE Americas, Belo Horizonte 30190-002, Brazil
| |
Collapse
|
10
|
Lozano-Parra A, Herrera V, Calderón C, Badillo R, Gélvez Ramírez RM, Estupiñán Cárdenas MI, Lozano Jiménez JF, Villar LÁ, Rojas Garrido EM. Chronic Rheumatologic Disease in Chikungunya Virus Fever: Results from a Cohort Study Conducted in Piedecuesta, Colombia. Trop Med Infect Dis 2024; 9:247. [PMID: 39453274 PMCID: PMC11511048 DOI: 10.3390/tropicalmed9100247] [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: 09/16/2024] [Revised: 10/10/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
This study aimed to determine the incidence of post-chikungunya chronic rheumatism (pCHIK-CR) and its impact on quality of life (QoL) and chronic fatigue in adults seven years after the 2014-2015 CHIKV outbreak in Piedecuesta, Colombia. We evaluated 78 adults (median age: 30 years, IQR: 21.0; women 60.3%) with confirmed CHIKV infection. In 2022, participants underwent a GALS examination and completed surveys on disability, stiffness, health status, and fatigue. A rheumatologist evaluated patients who reported arthralgia, morning stiffness, and abnormal GALS examination. Chronic fatigue was defined as fatigue persisting for over six months. Seven years after infection, 14.1% of participants were classified as pCHIK-CR cases, 41.0% as having non-inflammatory pain, likely degenerative (NIP-LD), and 44.9% without rheumatic disease (Wo-RM). Patients with pCHIK-CR and NIP-LD exhibited significantly worse QoL compared to Wo-RM cases. Chronic fatigue prevalence increased from 8.6% in Wo-RM patients to 25.0% in NIP-LD and 54.6% in pCHIK-CR cases. This study implemented a comprehensive clinical assessment to objectively estimate and characterize the incidence of chronic rheumatological disease attributed to CHIKV infection. One in seven cases with CHIKV infection develops pCHIK-CR, which impacts both QoL and chronic fatigue. This study contributes to understanding the burden of these arboviruses in the medium term.
Collapse
Affiliation(s)
- Anyela Lozano-Parra
- Grupo Epidemiología Clínica, Escuela de Medicina, Universidad Industrial de Santander UIS, Calle 9 Carrera 27, Bucaramanga 680002, Colombia; (A.L.-P.); (V.H.)
| | - Víctor Herrera
- Grupo Epidemiología Clínica, Escuela de Medicina, Universidad Industrial de Santander UIS, Calle 9 Carrera 27, Bucaramanga 680002, Colombia; (A.L.-P.); (V.H.)
| | - Carlos Calderón
- Centro de Atención y Diagnóstico de Enfermedades Infecciosas (CDI), Fundación INFOVIDA, Cra. 37 No. 51-126, Bucaramanga 680003, Colombia; (C.C.); (R.M.G.R.); (M.I.E.C.); (J.F.L.J.); (L.Á.V.)
| | - Reynaldo Badillo
- Departamento Medicina Interna, Universidad de Santander-UDES, Calle 35 # 10-43, Bucaramanga 680006, Colombia;
| | - Rosa Margarita Gélvez Ramírez
- Centro de Atención y Diagnóstico de Enfermedades Infecciosas (CDI), Fundación INFOVIDA, Cra. 37 No. 51-126, Bucaramanga 680003, Colombia; (C.C.); (R.M.G.R.); (M.I.E.C.); (J.F.L.J.); (L.Á.V.)
| | - María Isabel Estupiñán Cárdenas
- Centro de Atención y Diagnóstico de Enfermedades Infecciosas (CDI), Fundación INFOVIDA, Cra. 37 No. 51-126, Bucaramanga 680003, Colombia; (C.C.); (R.M.G.R.); (M.I.E.C.); (J.F.L.J.); (L.Á.V.)
| | - José Fernando Lozano Jiménez
- Centro de Atención y Diagnóstico de Enfermedades Infecciosas (CDI), Fundación INFOVIDA, Cra. 37 No. 51-126, Bucaramanga 680003, Colombia; (C.C.); (R.M.G.R.); (M.I.E.C.); (J.F.L.J.); (L.Á.V.)
| | - Luis Ángel Villar
- Centro de Atención y Diagnóstico de Enfermedades Infecciosas (CDI), Fundación INFOVIDA, Cra. 37 No. 51-126, Bucaramanga 680003, Colombia; (C.C.); (R.M.G.R.); (M.I.E.C.); (J.F.L.J.); (L.Á.V.)
| | - Elsa Marina Rojas Garrido
- Centro de Atención y Diagnóstico de Enfermedades Infecciosas (CDI), Fundación INFOVIDA, Cra. 37 No. 51-126, Bucaramanga 680003, Colombia; (C.C.); (R.M.G.R.); (M.I.E.C.); (J.F.L.J.); (L.Á.V.)
| |
Collapse
|
11
|
Brito RMDM, de Melo MF, Fernandes JV, Valverde JG, Matta Guedes PM, de Araújo JMG, Nascimento MSL. Acute Chikungunya Virus Infection Triggers a Diverse Range of T Helper Lymphocyte Profiles. Viruses 2024; 16:1387. [PMID: 39339863 PMCID: PMC11437511 DOI: 10.3390/v16091387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Chikungunya virus (CHIKV) is an arbovirus causing acute febrile illness with severe joint pain, often leading to chronic arthralgia. This study investigated the adaptive immune responses during the early stages of symptomatic acute CHIKV infection, focusing on the transcription factors and cytokines linked to Th1, Th2, Th17, and Treg cells. Thirty-six individuals were enrolled: nine healthy controls and 27 CHIKV-positive patients confirmed by qRT-PCR. Blood samples were analyzed for the mRNA expression of transcription factors (Tbet, GATA3, FoxP3, STAT3, RORγt) and cytokines (IFN-γ, IL-4, IL-17, IL-22, TGF-β, IL-10). The results showed the significant upregulation of Tbet, GATA3, FoxP3, STAT3, and RORγt in CHIKV-positive patients, with RORγt displaying the highest increase. Correspondingly, cytokines IFN-γ, IL-4, IL-17, and IL-22 were upregulated, while TGF-β was downregulated. Principal component analysis (PCA) confirmed the distinct immune profiles between CHIKV-positive and healthy individuals. A correlation analysis indicated that higher Tbet expression correlated with a lower viral load, whereas FoxP3 and TGF-β were associated with higher viral loads. Our study sheds light on the intricate immune responses during acute CHIKV infection, characterized by a mixed Th1, Th2, Th17, and Treg response profile. These results emphasize the complex interplay between different adaptive immune responses and how they may contribute to the pathogenesis of Chikungunya fever.
Collapse
Affiliation(s)
| | - Marília Farias de Melo
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - José Veríssimo Fernandes
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Joanna Gardel Valverde
- Institute of Tropical Medicine of Rio Grande do Norte, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Paulo Marcos Matta Guedes
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Josélio Maria Galvão de Araújo
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| | - Manuela Sales Lima Nascimento
- Department of Microbiology and Parasitology, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, Brazil
| |
Collapse
|
12
|
de Oliveira Souza R, Duarte Júnior JWB, Della Casa VS, Santoro Rosa D, Renia L, Claser C. Unraveling the complex interplay: immunopathology and immune evasion strategies of alphaviruses with emphasis on neurological implications. Front Cell Infect Microbiol 2024; 14:1421571. [PMID: 39211797 PMCID: PMC11358129 DOI: 10.3389/fcimb.2024.1421571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/09/2024] [Indexed: 09/04/2024] Open
Abstract
Arthritogenic alphaviruses pose a significant public health concern due to their ability to cause joint inflammation, with emerging evidence of potential neurological consequences. In this review, we examine the immunopathology and immune evasion strategies employed by these viruses, highlighting their complex mechanisms of pathogenesis and neurological implications. We delve into how these viruses manipulate host immune responses, modulate inflammatory pathways, and potentially establish persistent infections. Further, we explore their ability to breach the blood-brain barrier, triggering neurological complications, and how co-infections exacerbate neurological outcomes. This review synthesizes current research to provide a comprehensive overview of the immunopathological mechanisms driving arthritogenic alphavirus infections and their impact on neurological health. By highlighting knowledge gaps, it underscores the need for research to unravel the complexities of virus-host interactions. This deeper understanding is crucial for developing targeted therapies to address both joint and neurological manifestations of these infections.
Collapse
Affiliation(s)
- Raquel de Oliveira Souza
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | | | - Victória Simões Della Casa
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Daniela Santoro Rosa
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - Laurent Renia
- ASTAR Infectious Diseases Labs (ASTAR ID Labs), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Carla Claser
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
13
|
Flandes X, Hansen CA, Palani S, Abbas K, Bennett C, Caro WP, Hutubessy R, Khazhidinov K, Lambach P, Maure C, Marshall C, Rojas DP, Rosewell A, Sahastrabuddhe S, Tufet M, Wilder-Smith A, Beasley DWC, Bourne N, Barrett ADT. Vaccine value profile for Chikungunya. Vaccine 2024; 42:S9-S24. [PMID: 38407992 PMCID: PMC11554007 DOI: 10.1016/j.vaccine.2023.07.069] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 07/05/2023] [Accepted: 07/28/2023] [Indexed: 02/28/2024]
Abstract
Chikungunya virus (CHIKV) a mosquito-borne alphavirus is the causative agent of Chikungunya (CHIK), a disease with low mortality but high acute and chronic morbidity resulting in a high overall burden of disease. After the acute disease phase, chronic disease including persistent arthralgia is very common, and can cause fatigue and pain that is severe enough to limit normal activities. On average, around 40% of people infected with CHIKV will develop chronic arthritis, which may last for months or years. Recommendations for protection from CHIKV focus on infection control through preventing mosquito proliferation. There is currently no licensed antiviral drug or vaccine against CHIKV. Therefore, one of the most important public health impacts of vaccination would be to decrease burden of disease and economic losses in areas impacted by the virus, and prevent or reduce chronic morbidity associated with CHIK. This benefit would particularly be seen in Low and Middle Income Countries (LMIC) and socio-economically deprived areas, as they are more likely to have more infections and more severe outcomes. This 'Vaccine Value Profile' (VVP) for CHIK is intended to provide a high-level, holistic assessment of the information and data that are currently available to inform the potential public health, economic and societal value of vaccines in the development pipeline and vaccine-like products.This VVP was developed by a working group of subject matter experts from academia, non-profit organizations, public private partnerships, and multi-lateral organizations. All contributors have extensive expertise on various elements of the CHIK VVP and collectively aimed to identify current research and knowledge gaps.The VVP was developed using only existing and publicly available information.
Collapse
Affiliation(s)
- Ximena Flandes
- Department of Preventative Medicine and Population Health and University of Texas Medical Branch, Galveston, TX, United States
| | - Clairissa A Hansen
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Sunil Palani
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Kaja Abbas
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom; School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | | | | | | | | | | | - Clara Maure
- International Vaccine Institute, Seoul, Republic of Korea
| | | | | | | | | | - Marta Tufet
- Gavi the Vaccine Alliance, Geneva, Switzerland
| | | | - David W C Beasley
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, United States.
| | - Nigel Bourne
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, United States; Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States.
| | - Alan D T Barrett
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States; Department of Pediatrics, University of Texas Medical Branch, Galveston, TX, United States.
| |
Collapse
|
14
|
Shaikh MS, Faiyazuddin M, Khan MS, Pathan SK, Syed IJ, Gholap AD, Akhtar MS, Sah R, Mehta R, Sah S, Bonilla-Aldana DK, Luna C, Rodriguez-Morales AJ. Chikungunya virus vaccine: a decade of progress solving epidemiological dilemma, emerging concepts, and immunological interventions. Front Microbiol 2024; 15:1413250. [PMID: 39104592 PMCID: PMC11298817 DOI: 10.3389/fmicb.2024.1413250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Chikungunya virus (CHIKV), a single-stranded RNA virus transmitted by Aedes mosquitoes, poses a significant global health threat, with severe complications observed in vulnerable populations. The only licensed vaccine, IXCHIQ, approved by the US FDA, is insufficient to address the growing disease burden, particularly in endemic regions lacking herd immunity. Monoclonal antibodies (mAbs), explicitly targeting structural proteins E1/E2, demonstrate promise in passive transfer studies, with mouse and human-derived mAbs showing protective efficacy. This article explores various vaccine candidates, including live attenuated, killed, nucleic acid-based (DNA/RNA), virus-like particle, chimeric, subunit, and adenovirus vectored vaccines. RNA vaccines have emerged as promising candidates due to their rapid response capabilities and enhanced safety profile. This review underscores the importance of the E1 and E2 proteins as immunogens, emphasizing their antigenic potential. Several vaccine candidates, such as CHIKV/IRES, measles vector (MV-CHIK), synthetic DNA-encoded antibodies, and mRNA-lipid nanoparticle vaccines, demonstrate encouraging preclinical and clinical results. In addition to identifying potential molecular targets for antiviral therapy, the study looks into the roles played by Toll-like receptors, RIG-I, and NOD-like receptors in the immune response to CHIKV. It also offers insights into novel tactics and promising vaccine candidates. This article discusses potential antiviral targets, the significance of E1 and E2 proteins, monoclonal antibodies, and RNA vaccines as prospective Chikungunya virus vaccine candidates.
Collapse
Affiliation(s)
| | - Md. Faiyazuddin
- School of Pharmacy, Al – Karim University, Katihar, India
- Centre for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | | | - Shahbaz K. Pathan
- Medmecs Medical Coding & Billing Services, Universal Business Park, Mumbai, Maharashtra, India
| | - Imran J. Syed
- Y. B. Chavan College of Pharmacy, Aurangabad, Maharashtra, India
- SBSPM’s B. Pharmacy College, Beed, Maharashtra, India
| | - Amol D. Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar, Maharashtra, India
| | - Mohammad Shabib Akhtar
- Department of Clinical Pharmacy, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Ranjit Sah
- Green City Hospital, Kathmandu, Nepal
- Research Unit, Department of Microbiology, Dr. DY Patil Medical College, Hospital and Research Centre, DY Patil Vidyapeeth, Pune, Maharashtra, India
- Department of Public Health Dentistry, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Rachana Mehta
- Dr Lal PathLabs Nepal, Kathmandu, Nepal
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
- Clinical Microbiology, School of Dental Science, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana, India
| | | | | | - Camila Luna
- Faculty of Health Sciences, Universidad Científica del Sur, Lima, Peru
| | - Alfonso J. Rodriguez-Morales
- Faculty of Health Sciences, Universidad Científica del Sur, Lima, Peru
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundación Universitaria Autónoma de las Américas-Institución Universitaria Visión de las Américas, Pereira, Colombia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| |
Collapse
|
15
|
Silveira-Freitas JEP, Campagnolo ML, dos Santos Cortez M, de Melo FF, Zarpelon-Schutz AC, Teixeira KN. Long chikungunya? An overview to immunopathology of persistent arthralgia. World J Virol 2024; 13:89985. [PMID: 38984075 PMCID: PMC11229846 DOI: 10.5501/wjv.v13.i2.89985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/09/2024] [Accepted: 04/12/2024] [Indexed: 06/24/2024] Open
Abstract
Chikungunya fever (CF) is caused by an arbovirus whose manifestations are extremely diverse, and it has evolved with significant severity in recent years. The clinical signs triggered by the Chikungunya virus are similar to those of other arboviruses. Generally, fever starts abruptly and reaches high levels, followed by severe polyarthralgia and myalgia, as well as an erythematous or petechial maculopapular rash, varying in severity and extent. Around 40% to 60% of affected individuals report persistent arthralgia, which can last from months to years. The symptoms of CF mainly represent the tissue tropism of the virus rather than the immunopathogenesis triggered by the host's immune system. The main mechanisms associated with arthralgia have been linked to an increase in T helper type 17 cells and a consequent increase in receptor activator of nuclear factor kappa-Β ligand and bone resorption. This review suggests that persistent arthralgia results from the presence of viral antigens post-infection and the constant activation of signaling lymphocytic activation molecule family member 7 in synovial macrophages, leading to local infiltration of CD4+ T cells, which sustains the inflammatory process in the joints through the secretion of pro-inflammatory cytokines. The term "long chikungunya" was used in this review to refer to persistent arthralgia since, due to its manifestation over long periods after the end of the viral infection, this clinical condition seems to be characterized more as a sequel than as a symptom, given that there is no active infection involved.
Collapse
Affiliation(s)
| | | | | | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Campus Anísio Teixeira, Vitória da Conquista, Bahia 45029-094, Brazil
| | - Ana Carla Zarpelon-Schutz
- Campus Toledo, Universidade Federal do Paraná, Toledo, Paraná 85919-899, Brazil
- Programa de Pós-graduação em Biotecnologia, Palotina, Universidade Federal do Paraná-Setor Palotina, Paraná 85950-000, Brazil
| | - Kádima Nayara Teixeira
- Campus Toledo, Universidade Federal do Paraná, Toledo, Paraná 85919-899, Brazil
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular, Palotina, Universidade Federal do Paraná-Setor Palotina, Paraná 85950-000, Brazil
| |
Collapse
|
16
|
Rawat SK, Kale D, Nema S, Nema RK, Gupta S, Khadanga S, Biswas D. Detectable Viremia at Presentation Is a Predictor of Disease Severity in Chikungunya. Cureus 2024; 16:e58188. [PMID: 38741833 PMCID: PMC11089836 DOI: 10.7759/cureus.58188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2024] [Indexed: 05/16/2024] Open
Abstract
Background Chikungunya is a mosquito-borne re-emerging disease that has caused a significant number of outbreaks recently in diverse geographic settings across the globe. It leads to severe debilitating illness in a significant proportion of persons who are infected. Measures to limit the impact produced by recurrent outbreaks of the disease are limited and there is an urgent clinical need for early identification of those predisposed to develop severe disease. A comprehensive understanding regarding the proportion of individuals predisposed to developing severe disease is lacking as its correlation with detectable viremia is hinted at by some studies. In this context, we hypothesized that detectable viremia reflected in the diagnostic RT-PCR assay could be significantly associated with the development of severe disease in Chikungunya among those diagnosed on the basis of seroconversion. Our study aims to confirm the same in relation to disease severity among the suspected patients of Chikungunya in the setting of a tertiary care center. Methods In a prospective observational study at a tertiary care center, a total number of 1021 Chikungunya suspects presenting within seven days of illness were screened with Chikungunya Virus IgM ELISA from 2021 to 2023. Those having positive IgM results were further tested with RT-PCR in a blinded manner. According to the information entered into the predesigned form and the hospital follow-up/discharge data, the cases where symptoms like fever and joint pain persisted beyond two weeks were classified as severe versus those resolving within two weeks as mild. The patients in each group were compared for their clinical symptoms and association with the disease severity with detectable viremia (RT-PCR positivity). Results We identified a total of 178 (17.4%) lab-confirmed Chikungunya IgM-positive cases amongst the recruited patients. Here a total of 31 (18.9%) cases could be classified as severe and 133 (74.7%) as mild illness, the remaining 14 patients were excluded from analysis due to insufficient clinical data. Severe illness was significantly higher in elderly individuals belonging to more than 60 years (p = 0.01). Viremia was detected in 16 (9%), those with detectable viremia had higher odds (OR = 4.1) of manifesting as severe disease. Among the severe cases, the proportion of cases with RT-PCR positivity (8, 25.8%) at presentation was significantly higher (P = 0.01) versus those who presented with mild disease (7, 5.5%). Conclusion Our study reveals a correlation between detectable viremia in Chikungunya virus (CHIKV) patients and an increased risk of manifesting into a severe disease, where severe cases exhibited a significantly higher proportion of viremia, indicated by RT-PCR positivity. This study hints at the presence of viremia, joint symptoms, and elderly age as potentially useful clinical predictors of disease outcomes, these may serve as indicators for closer monitoring among individuals seeking medical attention due to Chikungunya infection. However, we need to validate these findings in future longitudinal studies incorporating multiple, time-bound follow-up data on clinical outcomes, viral titers, and its long-term complications.
Collapse
Affiliation(s)
- Sumit K Rawat
- Microbiology, Bundelkhand Medical College, Sagar, IND
- Microbiology, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| | - Dipesh Kale
- Microbiology, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| | - Shashwati Nema
- Microbiology, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| | - Ram K Nema
- Molecular Biology, Indian Council of Medical Research (ICMR) National Institute for Research in Environmental Health, Bhopal, IND
| | - Sudheer Gupta
- Next-Generation Sequencing (NGS) & Bioinformatics Division, 3B Blackbio Dx Ltd., Bhopal, IND
| | - Sagar Khadanga
- Internal Medicine, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| | - Debasis Biswas
- Microbiology, All India Institute of Medical Sciences, Bhopal, Bhopal, IND
| |
Collapse
|
17
|
Hernández-Sarmiento LJ, Tamayo-Molina YS, Valdés-López JF, Urcuqui-Inchima S. Mayaro virus infection elicits a robust pro-inflammatory and antiviral response in human macrophages. Acta Trop 2024; 252:107146. [PMID: 38342287 DOI: 10.1016/j.actatropica.2024.107146] [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: 09/04/2023] [Revised: 02/07/2024] [Accepted: 02/07/2024] [Indexed: 02/13/2024]
Abstract
Mayaro virus (MAYV), the etiological agent of Mayaro fever (MAYF), is an emergent arbovirus pathogen belonging to Togaviridae family. MAYF is characterized by high inflammatory component that can cause long-lasting arthralgia that persists for months. Macrophages are viral targets and reservoirs, key components of innate immunity and host response. Given the importance of this pathogen, our aim was to determine the inflammatory and antiviral response of human monocyte-derived macrophages (MDMs) infected with MAYV. First, we established the replication kinetics of the virus. Thereafter, we determined the expression of pattern recognition receptors, NF-ĸB complex, interferons (IFNs), two interleukin 27 (IL27) subunits, IFN-stimulated genes (ISGs), and the production of cytokines/chemokines. We found that human MDMs are susceptible to MAYV infection in vitro, with a peak of viral particles released between 24- and 48-hours post-infection (h.p.i) at MOI 0.5, and between 12 and 24 h.p.i at MOI 1. Interestingly, we observed a significant decline in the production of infectious viral particles at 72 h.p.i that was associated with the induction of antiviral response and high cytotoxic effect of MAYV infection in MDMs. We observed modulation of several genes after MAYV infection, as well, we noted the activation of antiviral detection and response pathways (Toll-like receptors, RIG-I/MDA5, and PKR) at 48 h.p.i but not at 6 h.p.i. Furthermore, MAYV-infected macrophages express high levels of the three types of IFNs and the two IL27 subunits at 48 h.p.i. Moreover, we found higher production of IL6, IL1β, CXCL8/IL8, CCL2, and CCL5 at 48 h.p.i as compared to 6 h.p.i. A robust antiviral response (ISG15, APOBEC3A, IFITM1, and MX2) was observed at 48 but not at 6 h.p.i. The innate and antiviral responses of MAYV-infected MDMs differ at 6 and 48 h.p.i. We conclude that MAYV infection induces robust pro-inflammatory and antiviral responses in human primary macrophages.
Collapse
Affiliation(s)
| | - Y S Tamayo-Molina
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Juan Felipe Valdés-López
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
18
|
Chang AY, Tritsch SR, Herrera Gomez CA, Encinales L, Cadena Bonfanti A, Rosales W, Mendoza-Torres E, Simmens S, Amdur RL, Mores CN, Fierbaugh P, Perez Hernandez CA, Avendaño G, Silvera PB, Crespo YG, Jimenez ADC, Martinez Zapata JC, Jimenez D, Osorio-Llanes E, Castellar-Lopez J, Suchowiecki K, Martins K, Gregory M, Zuluaga I, Proctor A, Hernández AS, Sierra-Carrero L, Colpas MV, Hernandez JCP, Quast AAF, De Barros JAC, Mejía JF, Ruiz JP, Boyle D, Firestein GS, Simon GL. Cytokine and T cell responses in post-chikungunya viral arthritis: A cross-sectional study. PLoS One 2024; 19:e0299521. [PMID: 38507338 PMCID: PMC10954186 DOI: 10.1371/journal.pone.0299521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 02/13/2024] [Indexed: 03/22/2024] Open
Abstract
OBJECTIVE To define the relationship between chronic chikungunya post-viral arthritis disease severity, cytokine response and T cell subsets in order to identify potential targets for therapy. METHODS Participants with chikungunya arthritis were recruited from Colombia from 2019-2021. Arthritis disease severity was quantified using the Disease Activity Score-28 and an Arthritis-Flare Questionnaire adapted for chikungunya arthritis. Plasma cytokine concentrations (interleukin (IL)-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13, interferon-γ and tumor necrosis factor (TNF)) were measured using a Meso Scale Diagnostics assay. Peripheral blood T cell subsets were measured using flow cytometry. RESULTS Among participants with chikungunya arthritis (N = 158), IL-2 levels and frequency of regulatory T cells (Tregs) were low. Increased arthritis disease activity was associated with higher levels of inflammatory cytokines (IL-6, TNF and CRP) and immunoregulatory cytokine IL-10 (p<0.05). Increased arthritis flare activity was associated with higher Treg frequencies (p<0.05) without affecting T effector (Teff) frequencies, Treg/Teff ratios and Treg subsets. Finally, elevated levels of IL-2 were correlated with increased Treg frequency, percent Tregs out of CD4+ T cells, and Treg subsets expressing immunosuppressive markers, while also correlating with an increased percent Teff out of live lymphocytes (p<0.05). CONCLUSION Chikungunya arthritis is characterized by increased inflammatory cytokines and deficient IL-2 and Treg responses. Greater levels of IL-2 were associated with improved Treg numbers and immunosuppressive markers. Future research may consider targeting these pathways for therapy.
Collapse
Affiliation(s)
- Aileen Y. Chang
- Department of Medicine, George Washington University, Washington, DC, United States of America
| | - Sarah R. Tritsch
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington, DC, United States of America
| | | | - Liliana Encinales
- Department of Medicine, Allied Research Society, Barranquilla, Atlántico, Colombia
| | | | - Wendy Rosales
- Advanced Biomedicine Research Group, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla, Atlántico, Colombia
| | - Evelyn Mendoza-Torres
- Advanced Biomedicine Research Group, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla, Atlántico, Colombia
| | - Samuel Simmens
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Washington, DC, United States of America
| | - Richard L. Amdur
- Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York, United States of America
| | - Christopher N. Mores
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington, DC, United States of America
| | - Paige Fierbaugh
- Department of Medicine, George Washington University, Washington, DC, United States of America
| | | | - Geraldine Avendaño
- Centro de Investigación, Clínica de la Costa SAS, Barranquilla, Atlántico, Colombia
| | - Paula Bruges Silvera
- Centro de Investigación, Clínica de la Costa SAS, Barranquilla, Atlántico, Colombia
| | | | | | | | - Dennys Jimenez
- Centro de Investigación, Clínica de la Costa SAS, Barranquilla, Atlántico, Colombia
| | - Estefanie Osorio-Llanes
- Advanced Biomedicine Research Group, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla, Atlántico, Colombia
| | - Jairo Castellar-Lopez
- Advanced Biomedicine Research Group, Universidad Libre de Colombia, Seccional Barranquilla, Barranquilla, Atlántico, Colombia
| | - Karol Suchowiecki
- Department of Medicine, George Washington University, Washington, DC, United States of America
| | - Karen Martins
- U.S. Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Melissa Gregory
- Henry M. Jackson Foundation, In Support of Austere Environments Consortium for Enhanced Sepsis Outcomes (ACESO), Bethesda, Maryland, United States of America
| | - Ivan Zuluaga
- Universidad Libre de Barranquilla, Clínica Iberoamérica, Barranquilla, Atlántico, Colombia
| | - Abigale Proctor
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington, DC, United States of America
| | | | | | | | | | | | | | - José Forero Mejía
- Centro de Investigación, Clínica de la Costa SAS, Barranquilla, Atlántico, Colombia
| | - Johan Penagos Ruiz
- Centro de Investigación, Clínica de la Costa SAS, Barranquilla, Atlántico, Colombia
| | - David Boyle
- Department of Medicine, UC San Diego School of Medicine, San Diego, CA, United States of America
| | - Gary S. Firestein
- Department of Medicine, UC San Diego School of Medicine, San Diego, CA, United States of America
| | - Gary L. Simon
- Department of Medicine, George Washington University, Washington, DC, United States of America
| |
Collapse
|
19
|
de Lima RC, Valente LMM, Familiar Macedo D, de-Oliveira-Pinto LM, dos Santos FB, Mazzei JL, Siani AC, Nunes PCG, de Azeredo EL. Antiviral and Virucidal Activities of Uncaria tomentosa (Cat's Claw) against the Chikungunya Virus. Viruses 2024; 16:369. [PMID: 38543735 PMCID: PMC10974475 DOI: 10.3390/v16030369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 05/23/2024] Open
Abstract
Uncaria tomentosa (UT) is a medicinal plant popularly known as cat's claw belonging to the Rubiaceae family that has been reported to display antiviral and anti-inflammatory activities. The chikungunya virus (CHIKV) outbreaks constitute a Brazilian public health concern. CHIKV infection develops an abrupt onset of fever, usually accompanied by a skin rash, besides incapacitating polyarthralgia. There is no vaccine available or treatment for CHIKV infection. The present study evaluates the hydroalcoholic extract of UT bark as a potential antiviral against CHIKV. The in vitro antiviral activity of the UT extract against the Brazilian CHIKV strain was assessed using quantitative reverse transcription polymerase chain reaction, flow cytometry, and plaque assay. Results obtained demonstrated that UT inhibits CHIKV infection in a dose-dependent manner. At the non-cytotoxic concentration of 100 µg/mL, UT exhibited antiviral activity above 90% as determined by plaque reduction assay, and it reduced the viral cytopathic effect. Similarly, a significant virucidal effect of 100 µg/mL UT was observed after 24 and 48 h post-infection. This is the first report on the antiviral activity of UT against CHIKV infection, and the data presented here suggests UT as a potential antiviral to treat CHIKV infection.
Collapse
Affiliation(s)
- Raquel Curtinhas de Lima
- Laboratório das Interações Vírus Hospedeiros, Instituto Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil; (R.C.d.L.); (D.F.M.); (L.M.d.-O.-P.); (F.B.d.S.); (P.C.G.N.)
| | - Ligia Maria Marino Valente
- Instituto de Química, Universidade Federal do Rio de Janeiro, Av. Athos da Silveira Ramos, 149, Rio de Janeiro 21941-909, Brazil;
| | - Débora Familiar Macedo
- Laboratório das Interações Vírus Hospedeiros, Instituto Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil; (R.C.d.L.); (D.F.M.); (L.M.d.-O.-P.); (F.B.d.S.); (P.C.G.N.)
| | - Luzia Maria de-Oliveira-Pinto
- Laboratório das Interações Vírus Hospedeiros, Instituto Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil; (R.C.d.L.); (D.F.M.); (L.M.d.-O.-P.); (F.B.d.S.); (P.C.G.N.)
| | - Flavia Barreto dos Santos
- Laboratório das Interações Vírus Hospedeiros, Instituto Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil; (R.C.d.L.); (D.F.M.); (L.M.d.-O.-P.); (F.B.d.S.); (P.C.G.N.)
| | - José Luiz Mazzei
- Laboratório de Tecnologia para Biodiversidade em Saúde, Instituto de Tecnologia de Fármacos, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, Brazil; (J.L.M.); (A.C.S.)
| | - Antonio Carlos Siani
- Laboratório de Tecnologia para Biodiversidade em Saúde, Instituto de Tecnologia de Fármacos, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, Brazil; (J.L.M.); (A.C.S.)
| | - Priscila Conrado Guerra Nunes
- Laboratório das Interações Vírus Hospedeiros, Instituto Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil; (R.C.d.L.); (D.F.M.); (L.M.d.-O.-P.); (F.B.d.S.); (P.C.G.N.)
| | - Elzinandes Leal de Azeredo
- Laboratório das Interações Vírus Hospedeiros, Instituto Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil; (R.C.d.L.); (D.F.M.); (L.M.d.-O.-P.); (F.B.d.S.); (P.C.G.N.)
| |
Collapse
|
20
|
Atella MO, Carvalho AS, Da Poian AT. Role of macrophages in the onset, maintenance, or control of arthritis caused by alphaviruses. Exp Biol Med (Maywood) 2023; 248:2039-2044. [PMID: 38058027 PMCID: PMC10800133 DOI: 10.1177/15353702231214261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023] Open
Abstract
Arthritogenic alphaviruses are mosquito-borne viruses that cause a debilitating rheumatic disease characterized by fever, headache, rash, myalgia, and polyarthralgia with the potential to evolve into a severe and very prolonged illness. Although these viruses have been geographically restricted by vector hosts and reservoirs, recent epidemics have revealed the risks of their spread worldwide. In this review, we aim to discuss the protective and pathological roles of macrophages during the development of arthritis caused by alphaviruses. The progression to the chronic phase of the disease is related to the extension of viral replication and the maintenance of articular inflammation, in which the cellular infiltrate is predominantly composed of macrophages. We explore the possible implications of macrophage polarization to M1/M2 activation phenotypes, drawing a parallel between alphavirus arthritis and rheumatoid arthritis (RA), a chronic inflammatory disease that also affects articular tissues. In RA, it is well established that M1 macrophages contribute to tissue damage and inflammation, while M2 macrophages have a role in cartilage repair, so modulating the M1/M2 macrophage ratio is being considered as a strategy in the treatment of this disease. In the case of alphavirus-induced arthritis, the picture is more complex, as proinflammatory factors derived from M1 macrophages contribute to the antiviral response but cause tissue damage, while M2 macrophages may contribute to tissue repair but impair viral clearance.
Collapse
Affiliation(s)
| | | | - Andrea T Da Poian
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| |
Collapse
|
21
|
Ravindran S, Lahon A. Tropism and immune response of chikungunya and zika viruses: An overview. Cytokine 2023; 170:156327. [PMID: 37579710 DOI: 10.1016/j.cyto.2023.156327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/16/2023]
Abstract
Zika virus (ZIKV) and chikungunya virus (CHIKV) are two medically important vector-borne viruses responsible for causing significant disease burden in humans, including neurological sequelae/complications. Besides sharing some common clinical features, ZIKV has major shares in causing microcephaly and brain malformations in developing foetus, whereas CHIKV causes chronic joint pain/swelling in infected individuals. Both viruses have a common route of entry to the host body. i.e., dermal site of inoculation through the bite of an infected mosquito and later taken up by different immune cells for further dissemination to other areas of the host body that lead to a range of immune responses via different pathways. The immune responses generated by both viruses have similar characteristics with varying degrees of inflammation and activation of immune cells. However, the overall response of immune cells is not fully explored in the context of ZIKV and CHIKV infection. The knowledge of cellular tropism and the immune response is the key to understanding the mechanisms of viral immunity and pathogenesis, which may allow to develop novel therapeutic strategies for these viral infections. This review aims to discuss recent advancements and identify the knowledge gaps in understanding the mechanism of cellular tropism and immune response of CHIKV and ZIKV.
Collapse
Affiliation(s)
- Shilpa Ravindran
- Institute of Advanced Virology, Thiruvananthapuram, Kerala 695317, India
| | - Anismrita Lahon
- Institute of Advanced Virology, Thiruvananthapuram, Kerala 695317, India.
| |
Collapse
|
22
|
Sandenon Seteyen AL, Guiraud P, Gasque P, Girard-Valenciennes E, Sélambarom J. In Vitro Analyses of the Multifocal Effects of Natural Alkaloids Berberine, Matrine, and Tabersonine against the O'nyong-nyong Arthritogenic Alphavirus Infection and Inflammation. Pharmaceuticals (Basel) 2023; 16:1125. [PMID: 37631040 PMCID: PMC10459185 DOI: 10.3390/ph16081125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
O'nyong-nyong virus (ONNV) is a member of the reemerging arthritogenic alphaviruses that cause chronic debilitating polyarthralgia and/or polyarthritis via their tropism for the musculoskeletal system. Thus, the discovery of dual antiviral and anti-inflammatory drugs is a great challenge in this field. We investigated the effects of the common plant-derived alkaloids berberine (isoquinoline), matrine (quinolizidine), and tabersonine (indole) at a non-toxic concentration (10 μM) on a human fibroblast cell line (HS633T) infected by ONNV (MOI 1). Using qRT-PCR analyses, we measured the RNA levels of the gene coding for the viral proteins and for the host cell immune factors. These alkaloids demonstrated multifocal effects by the inhibition of viral replication, as well as the regulation of the type-I interferon antiviral signaling pathway and the inflammatory mediators and pathways. Berberine and tabersonine proved to be the more valuable compounds. The results supported the proposal that these common alkaloids may be useful scaffolds for drug discovery against arthritogenic alphavirus infection.
Collapse
Affiliation(s)
- Anne-Laure Sandenon Seteyen
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France; (A.-L.S.S.); (P.G.)
| | - Pascale Guiraud
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France; (A.-L.S.S.); (P.G.)
| | - Philippe Gasque
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France; (A.-L.S.S.); (P.G.)
- Centre Hospitalier Universitaire de La Réunion, Laboratoire d’Immunologie Clinique et Expérimentale de la Zone Océan Indien (LICE-OI), Pôle de Biologie, 97400 Saint-Denis, France
| | - Emmanuelle Girard-Valenciennes
- Laboratoire de Chimie et de Biotechnologie des Produits Naturels (CHEMBIOPRO), Université de La Réunion, 97400 Saint-Denis, France
| | - Jimmy Sélambarom
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France; (A.-L.S.S.); (P.G.)
| |
Collapse
|
23
|
Barker D, Han X, Wang E, Dagley A, Anderson DM, Jha A, Weaver SC, Julander J, Nykiforuk C, Kodihalli S. Equine Polyclonal Antibodies Prevent Acute Chikungunya Virus Infection in Mice. Viruses 2023; 15:1479. [PMID: 37515166 PMCID: PMC10384969 DOI: 10.3390/v15071479] [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: 05/08/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted pathogen that causes chikungunya disease (CHIK); the disease is characterized by fever, muscle ache, rash, and arthralgia. This arthralgia can be debilitating and long-lasting, seriously impacting quality of life for years. Currently, there is no specific therapy available for CHIKV infection. We have developed a despeciated equine polyclonal antibody (CHIKV-EIG) treatment against CHIKV and evaluated its protective efficacy in mouse models of CHIKV infection. In immunocompromised (IFNAR-/-) mice infected with CHIKV, daily treatment for five consecutive days with CHIKV-EIG administered at 100 mg/kg starting on the day of infection prevented mortality, reduced viremia, and improved clinical condition as measured by body weight loss. These beneficial effects were seen even when treatment was delayed to 1 day after infection. In immunocompetent mice, CHIKV-EIG treatment reduced virus induced arthritis (including footpad swelling), arthralgia-associated cytokines, viremia, and tissue virus loads in a dose-dependent fashion. Collectively, these results suggest that CHIKV-EIG is effective at preventing CHIK and could be a viable candidate for further development as a treatment for human disease.
Collapse
Affiliation(s)
- Douglas Barker
- Emergent BioSolutions Canada Inc., Winnipeg, MB R3T 5Y3, Canada
| | - Xiaobing Han
- Emergent BioSolutions Canada Inc., Winnipeg, MB R3T 5Y3, Canada
| | - Eryu Wang
- Institute for Human Infections and Immunity, Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, Galveston, TX 77555, USA
| | - Ashley Dagley
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | | | - Aruni Jha
- Emergent BioSolutions Canada Inc., Winnipeg, MB R3T 5Y3, Canada
| | - Scott C Weaver
- Institute for Human Infections and Immunity, Department of Microbiology and Immunology, University of Texas Medical Branch Galveston, Galveston, TX 77555, USA
| | - Justin Julander
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Cory Nykiforuk
- Emergent BioSolutions Canada Inc., Winnipeg, MB R3T 5Y3, Canada
| | | |
Collapse
|
24
|
Hossain MA, Sohel M, Sultana T, Hasan MI, Khan MS, Kibria KMK, Mahmud SMH, Rahman MH. Study of kaempferol in the treatment of COVID-19 combined with Chikungunya co-infection by network pharmacology and molecular docking technology. INFORMATICS IN MEDICINE UNLOCKED 2023; 40:101289. [PMID: 37346467 PMCID: PMC10264333 DOI: 10.1016/j.imu.2023.101289] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/23/2023] Open
Abstract
Chikungunya (CHIK) patients may be vulnerable to coronavirus disease (COVID-19). However, presently there are no anti-COVID-19/CHIK therapeutic alternatives available. The purpose of this research was to determine the pharmacological mechanism through which kaempferol functions in the treatment of COVID-19-associated CHIK co-infection. We have used a series of network pharmacology and computational analysis-based techniques to decipher and define the binding capacity, biological functions, pharmacological targets, and treatment processes in COVID-19-mediated CHIK co-infection. We identified key therapeutic targets for COVID-19/CHIK, including TP53, MAPK1, MAPK3, MAPK8, TNF, IL6 and NFKB1. Gene ontology, molecular and upstream pathway analysis of kaempferol against COVID-19 and CHIK showed that DEGs were confined mainly to the cytokine-mediated signalling pathway, MAP kinase activity, negative regulation of the apoptotic process, lipid and atherosclerosis, TNF signalling pathway, hepatitis B, toll-like receptor signaling, IL-17 and IL-18 signaling pathways. The study of the gene regulatory network revealed several significant TFs including KLF16, GATA2, YY1 and FOXC1 and miRNAs such as let-7b-5p, mir-16-5p, mir-34a-5p, and mir-155-5p that target differential-expressed genes (DEG). According to the molecular coupling results, kaempferol exhibited a high affinity for 5 receptor proteins (TP53, MAPK1, MAPK3, MAPK8, and TNF) compared to control inhibitors. In combination, our results identified significant targets and pharmacological mechanisms of kaempferol in the treatment of COVID-19/CHIK and recommended that core targets be used as potential biomarkers against COVID-19/CHIK viruses. Before conducting clinical studies for the intervention of COVID-19 and CHIK, kaempferol might be evaluated in wet lab tests at the molecular level.
Collapse
Affiliation(s)
- Md Arju Hossain
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail, 1902, Bangladesh
| | - Md Sohel
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Santosh, Tangail, 1902, Bangladesh
- Department of Biochemistry and Molecular Biology, Primeasia University, Dhaka, Bangladesh
| | - Tayeba Sultana
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail, 1902, Bangladesh
| | - Md Imran Hasan
- Department of Computer Science and Engineering, Islamic University, Kushtia, 7003, Bangladesh
| | - Md Sharif Khan
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail, 1902, Bangladesh
| | - K M Kaderi Kibria
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail, 1902, Bangladesh
| | - S M Hasan Mahmud
- Department of Computer Science, Faculty of Science and Technology, American International University-Bangladesh, Dhaka, Bangladesh
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Islamic University, Kushtia, 7003, Bangladesh
- Center for Advanced Bioinformatics and Artificial Intelligent Research, Islamic University, Kushtia, 7003, Bangladesh
| |
Collapse
|
25
|
Srivastava P, Chaudhary S, Malhotra S, Varma B, Sunil S. Transcriptome analysis of human macrophages upon chikungunya virus (CHIKV) infection reveals regulation of distinct signaling and metabolic pathways during the early and late stages of infection. Heliyon 2023; 9:e17158. [PMID: 37408916 PMCID: PMC10318463 DOI: 10.1016/j.heliyon.2023.e17158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 06/01/2023] [Accepted: 06/08/2023] [Indexed: 07/07/2023] Open
Abstract
Macrophages are efficient reservoirs for viruses that enable the viruses to survive over a longer period of infection. Alphaviruses such as chikungunya virus (CHIKV) are known to persist in macrophages even after the acute febrile phase. The viral particles replicate in macrophages at a very low level over extended period of time and are localized in tissues that are often less accessible by treatment. Comprehensive experimental studies are thus needed to characterize the CHIKV-induced modulation of host genes in these myeloid lineage cells and in one such pursuit, we obtained global transcriptomes of a human macrophage cell line infected with CHIKV, over its early and late timepoints of infection. We analyzed the pathways, especially immune related, perturbed over these timepoints and observed several host factors to be differentially expressed in infected macrophages in a time-dependent manner. We postulate that these pathways may play crucial roles in the persistence of CHIKV in macrophages.
Collapse
Affiliation(s)
- Priyanshu Srivastava
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Sakshi Chaudhary
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | | | | | - Sujatha Sunil
- Vector-Borne Diseases Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| |
Collapse
|
26
|
Henderson Sousa F, Ghaisani Komarudin A, Findlay-Greene F, Bowolaksono A, Sasmono RT, Stevens C, Barlow PG. Evolution and immunopathology of chikungunya virus informs therapeutic development. Dis Model Mech 2023; 16:dmm049804. [PMID: 37014125 PMCID: PMC10110403 DOI: 10.1242/dmm.049804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
Chikungunya virus (CHIKV), a mosquito-borne alphavirus, is an emerging global threat identified in more than 60 countries across continents. The risk of CHIKV transmission is rising due to increased global interactions, year-round presence of mosquito vectors, and the ability of CHIKV to produce high host viral loads and undergo mutation. Although CHIKV disease is rarely fatal, it can progress to a chronic stage, during which patients experience severe debilitating arthritis that can last from several weeks to months or years. At present, there are no licensed vaccines or antiviral drugs for CHIKV disease, and treatment is primarily symptomatic. This Review provides an overview of CHIKV pathogenesis and explores the available therapeutic options and the most recent advances in novel therapeutic strategies against CHIKV infections.
Collapse
Affiliation(s)
- Filipa Henderson Sousa
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh EH11 4BN, UK
- Centre for Discovery Brain Sciences and UK Dementia Research Institute, The University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Amalina Ghaisani Komarudin
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong Science Center, Cibinong, Kabupaten Bogor 16911, Indonesia
| | - Fern Findlay-Greene
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh EH11 4BN, UK
| | - Anom Bowolaksono
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Indonesia, Depok 16424, Indonesia
| | - R. Tedjo Sasmono
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong Science Center, Cibinong, Kabupaten Bogor 16911, Indonesia
| | - Craig Stevens
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh EH11 4BN, UK
| | - Peter G. Barlow
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh EH11 4BN, UK
| |
Collapse
|
27
|
Sengupta S, Bhattacharya N, Tripathi A. Increased CRP, anti-CCP antibody, IL-2R, COMP levels in prognosis of post-chikungunya chronic arthritis and protective role of their specific genotypes against arthritic manifestation. Virus Res 2023; 323:198998. [PMID: 36356677 PMCID: PMC10194367 DOI: 10.1016/j.virusres.2022.198998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/21/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022]
Abstract
Chikungunya infection leads to acute/chronic polyarthritis/polyarthralgia causing long-term morbidity among patients. Prognosis of post-chikungunya chronic arthritis (PCA) is of utmost necessity for proper disease management. Arthritic and hepatic biomarkers were evaluated among chikungunya patients without arthritis, with acute arthritis and with post-chikungunya chronic arthritis in the study. Serum levels of arthritic [CRP (C-reactive protein), anti cyclic-citrullinated-peptide (anti-CCP) antibody, soluble interleukin-2 receptor (sIL-2R), cartilage oligomeric matrix protein (COMP)] and hepatic [ALT (alanine aminotransferase), AST (aspartate aminotransferase), ALP (alkaline phosphatase), albumin and bilirubin] biomarkers of 167 chikungunya positive patients were determined by sandwich-ELISA/immunoturbidimetry/auto-analyser. 167 chikungunya-patients and 102 healthy controls were genotyped to understand role of CRP-rs3093059/rs3091244, IL-2R-rs743777 and COMP-rs144778694 polymorphisms towards chikungunya virus (CHIKV) infectivity and arthralgic manifestation. CRP, anti-CCP antibody, IL-2R and COMP levels significantly increased among PCA patients. Concentrations of AST, ALT, AST/ALT-ratio, bilirubin and ALP increased among arthritic chikungunya patients. Principal component analysis differentiated PCA groups from acute (AA) and non-arthritic groups. Patients with IL-2R-rs743777-GA, G-allele and COMP-rs144778694-GA genotypes were susceptible to chikungunya infection. Moreover, patients with CRP-rs3093059-CT, rs3091244-TT, IL-2R-rs743777-GA and COMP-rs144778694-AA genotypes were significantly protected from arthralgia, whereas, COMP-rs144778694-GA genotype was susceptible towards it. Patients with certain genotypes of CRP, IL-2R and COMP demonstrated significantly higher biomarker serum-levels among patients suffering from AA with/without PCA. Thus, both serum biomarker levels and polymorphic genotypes of infected patients play decisive role in development of PCA.
Collapse
Affiliation(s)
- Siddhartha Sengupta
- Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, 108, C. R. Avenue, Kolkata, West Bengal 700073, India
| | - Nemai Bhattacharya
- Department of Microbiology, Virus unit, Calcutta School of Tropical Medicine, 108, C. R. Avenue, Kolkata, West Bengal 700073, India
| | - Anusri Tripathi
- Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, 108, C. R. Avenue, Kolkata, West Bengal 700073, India.
| |
Collapse
|
28
|
Amaral JK, Bingham CO, Taylor PC, Vilá LM, Weinblatt ME, Schoen RT. Pathogenesis of chronic chikungunya arthritis: Resemblances and links with rheumatoid arthritis. Travel Med Infect Dis 2022; 52:102534. [PMID: 36549417 DOI: 10.1016/j.tmaid.2022.102534] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/03/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
Chikungunya virus (CHIKV) infection results from transmission by the mosquito vector. Following an incubation period of 5-7 days, patients develop an acute febrile illness, chikungunya fever (CHIKF), characterized by high fevers, maculopapular rash, headaches, polyarthritis/arthralgias, myalgias, nausea, vomiting, and diarrhea. Joint pain is often severe, and most often involves the hands, the wrists, the ankles, and the metatarsal-phalangeal joints of the feet. Many patients recover within several weeks, but up to 50% develop chronic joint pain and swelling for more than 12 weeks, then we refer to these symptoms as chronic chikungunya arthritis (CCA). The pathogenesis of CCA is not well understood. In this article, we suggest that mesenchymal stem cells (MSCs) may play an important role in this pathogenesis. This heterogeneous group of multipotent cells, morphologically similar to fibroblasts, may undergo epigenetic changes capable of generating aberrant progenies. However, we believe that there is no need for a latent infection. In our pathogenic hypothesis, CHIKV infection of MSCs would cause epigenetic changes both in MSCs themselves and in their progenies, without the need for reactivation of dormant viruses.
Collapse
Affiliation(s)
- J Kennedy Amaral
- Institute of Diagnostic Medicine of Cariri, Juazeiro do Norte, Ceará, Brazil.
| | - Clifton O Bingham
- Johns Hopkins Arthritis Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peter C Taylor
- University of Oxford, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Windmill Road, Oxford, UK
| | - Luis M Vilá
- Division of Rheumatology, Allergy and Immunology, San Juan, Puerto Rico, USA
| | - Michael E Weinblatt
- John R. and Eileen K. Riedman Professor of Medicine, Harvard Medical School, USA
| | - Robert T Schoen
- Section of Rheumatology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
29
|
Geng T, Yang D, Lin T, Cahoon JG, Wang P. UBXN3B Controls Immunopathogenesis of Arthritogenic Alphaviruses by Maintaining Hematopoietic Homeostasis. mBio 2022; 13:e0268722. [PMID: 36377866 PMCID: PMC9765034 DOI: 10.1128/mbio.02687-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
Ubiquitin regulatory X domain-containing proteins (UBXN) might be involved in diverse cellular processes. However, their in vivo physiological functions remain largely elusive. We recently showed that UBXN3B positively regulated stimulator-of-interferon-genes (STING)-mediated innate immune responses to DNA viruses. Herein, we reported the essential role of UBXN3B in the control of infection and immunopathogenesis of two arthritogenic RNA viruses, Chikungunya (CHIKV) and O'nyong'nyong (ONNV) viruses. Ubxn3b deficient (Ubxn3b-/-) mice presented higher viral loads, more severe foot swelling and immune infiltrates, and slower clearance of viruses and resolution of inflammation than the Ubxn3b+/+ littermates. While the serum cytokine levels were intact, the virus-specific immunoglobulin G and neutralizing antibody levels were lower in the Ubxn3b-/- mice. The Ubxn3b-/- mice had more neutrophils and macrophages, but much fewer B cells in the ipsilateral feet. Of note, this immune dysregulation was also observed in the spleens and blood of uninfected Ubxn3b-/- mice. UBXN3B restricted CHIKV replication in a cell-intrinsic manner but independent of type I IFN signaling. These results demonstrated a dual role of UBXN3B in the maintenance of immune homeostasis and control of RNA virus replication. IMPORTANCE The human genome encodes 13 ubiquitin regulatory X (UBX) domain-containing proteins (UBXN) that might participate in diverse cellular processes. However, their in vivo physiological functions remain largely elusive. Herein, we reported an essential role of UBXN3B in the control of infection and immunopathogenesis of arthritogenic alphaviruses, including Chikungunya virus (CHIKV), which causes acute and chronic crippling arthralgia, long-term neurological disorders, and poses a significant public health problem in the tropical and subtropical regions worldwide. However, there are no approved vaccines or specific antiviral drugs. This was partly due to a poor understanding of the protective and detrimental immune responses elicited by CHIKV. We showed that UBXN3B was critical for the control of CHIKV replication in a cell-intrinsic manner in the acute phase and persistent immunopathogenesis in the post-viremic stage. Mechanistically, UBXN3B was essential for the maintenance of hematopoietic homeostasis during viral infection and in steady-state.
Collapse
Affiliation(s)
- Tingting Geng
- Department of Immunology, School of Medicine, the University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Duomeng Yang
- Department of Immunology, School of Medicine, the University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Tao Lin
- Department of Immunology, School of Medicine, the University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Jason G. Cahoon
- Department of Immunology, School of Medicine, the University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Penghua Wang
- Department of Immunology, School of Medicine, the University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
30
|
Basavappa MG, Ferretti M, Dittmar M, Stoute J, Sullivan MC, Whig K, Shen H, Liu KF, Schultz DC, Beiting DP, Lynch KW, Henao-Mejia J, Cherry S. The lncRNA ALPHA specifically targets chikungunya virus to control infection. Mol Cell 2022; 82:3729-3744.e10. [PMID: 36167073 PMCID: PMC10464526 DOI: 10.1016/j.molcel.2022.08.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 07/06/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022]
Abstract
Arthropod-borne viruses, including the alphavirus chikungunya virus (CHIKV), cause acute disease in millions of people and utilize potent mechanisms to antagonize and circumvent innate immune pathways including the type I interferon (IFN) pathway. In response, hosts have evolved antiviral counterdefense strategies that remain incompletely understood. Recent studies have found that long noncoding RNAs (lncRNAs) regulate classical innate immune pathways; how lncRNAs contribute to additional antiviral counterdefenses remains unclear. Using high-throughput genetic screening, we identified a cytoplasmic antiviral lncRNA that we named antiviral lncRNA prohibiting human alphaviruses (ALPHA), which is transcriptionally induced by alphaviruses and functions independently of IFN to inhibit the replication of CHIKV and its closest relative, O'nyong'nyong virus (ONNV), but not other viruses. Furthermore, we showed that ALPHA interacts with CHIKV genomic RNA and restrains viral RNA replication. Together, our findings reveal that ALPHA and potentially other lncRNAs can mediate non-canonical antiviral immune responses against specific viruses.
Collapse
Affiliation(s)
- Megha G Basavappa
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Max Ferretti
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark Dittmar
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julian Stoute
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Megan C Sullivan
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kanupriya Whig
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA; High-Throughput Screening Core, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hui Shen
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathy Fange Liu
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David C Schultz
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA; High-Throughput Screening Core, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P Beiting
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristen W Lynch
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jorge Henao-Mejia
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Protective Immunity, Department of Pathology and Laboratory Medicine, Children's Hospital of Pennsylvania, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA; High-Throughput Screening Core, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
31
|
Gomes de Azevedo-Quintanilha I, Campos MM, Teixeira Monteiro AP, Dantas do Nascimento A, Calheiros AS, Oliveira DM, Dias SSG, Soares VC, Santos JDC, Tavares I, Lopes Souza TM, Hottz ED, Bozza FA, Bozza PT. Increased platelet activation and platelet-inflammasome engagement during chikungunya infection. Front Immunol 2022; 13:958820. [PMID: 36189282 PMCID: PMC9520464 DOI: 10.3389/fimmu.2022.958820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Chikungunya fever is a viral disease transmitted by mosquitoes of the genus Aedes. The infection is usually symptomatic and most common symptoms are fever accompanied by joint pain and swelling. In most cases symptoms subside within a week. However, severe prolonged and disabling joint pain, that may persist for several months, even years, are reported. Although the pathogenesis of Chikungunya infection is not fully understood, the evolution to severe disease seems to be associated with the activation of immune mechanisms and the action of inflammatory mediators. Platelets are recognized as inflammatory cells with fundamental activities in the immune response, maintenance of vascular stability and pathogenicity of several inflammatory and infectious diseases. Although the involvement of platelets in the pathogenesis of viral diseases has gained attention in recent years, their activation in Chikungunya has not been explored. The aim of this study was to analyze platelet activation and the possible role of platelets in the amplification of the inflammatory response during Chikungunya infection. We prospectively included 132 patients attended at the Quinta D’Or hospital and 25 healthy volunteers during the 2016 epidemic in Rio de Janeiro, Brazil. We observed increased expression of CD62P on the surface of platelets, as well as increased plasma levels of CD62P and platelet-derived inflammatory mediators indicating that the Chikungunya infection leads to platelet activation. In addition, platelets from chikungunya patients exhibit increased expression of NLRP3, caspase 4, and cleaved IL-1β, suggestive of platelet-inflammasome engagement during chikungunya infection. In vitro experiments confirmed that the Chikungunya virus directly activates platelets. Moreover, we observed that platelet activation and soluble p-selectin at the onset of symptoms were associated with development of chronic forms of the disease. Collectively, our data suggest platelet involvement in the immune processes and inflammatory amplification triggered by the infection.
Collapse
Affiliation(s)
- Isaclaudia Gomes de Azevedo-Quintanilha
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- *Correspondence: Patricia T. Bozza, ; Isaclaudia Gomes de Azevedo-Quintanilha,
| | - Mariana Macedo Campos
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Alessandra Dantas do Nascimento
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- Instituto D’Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
| | - Andrea Surrage Calheiros
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Douglas Mathias Oliveira
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Suelen Silva Gomes Dias
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Vinicius Cardoso Soares
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Julia da Cunha Santos
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Isabel Tavares
- Instituto D’Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
| | - Thiago Moreno Lopes Souza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- Centro de Desenvolvimento Tecnológico em Saúde (CDTS) and National Institute for Science and Technology on Innovation on Diseases of Neglected Populations (INCT/IDNP), FIOCRUZ, Rio de Janeiro, Brazil
| | - Eugenio D. Hottz
- Laboratório de Imunotrombose, Departamento de Bioquimica, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Fernando A. Bozza
- Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- Instituto D’Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
| | - Patricia T. Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- *Correspondence: Patricia T. Bozza, ; Isaclaudia Gomes de Azevedo-Quintanilha,
| |
Collapse
|
32
|
de Araújo S, de Melo Costa VR, Santos FM, de Sousa CDF, Moreira TP, Gonçalves MR, Félix FB, Queiroz-Junior CM, Campolina-Silva GH, Nogueira ML, Sugimoto MA, Bonilha CS, Perretti M, Souza DG, Costa VV, Teixeira MM. Annexin A1-FPR2/ALX Signaling Axis Regulates Acute Inflammation during Chikungunya Virus Infection. Cells 2022; 11:cells11172717. [PMID: 36078125 PMCID: PMC9454528 DOI: 10.3390/cells11172717] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 12/03/2022] Open
Abstract
Chikungunya (CHIKV) is an arthritogenic alphavirus that causes a self-limiting disease usually accompanied by joint pain and/or polyarthralgia with disabling characteristics. Immune responses developed during the acute phase of CHIKV infection determine the rate of disease progression and resolution. Annexin A1 (AnxA1) is involved in both initiating inflammation and preventing over-response, being essential for a balanced end of inflammation. In this study, we investigated the role of the AnxA1-FPR2/ALX pathway during CHIKV infection. Genetic deletion of AnxA1 or its receptor enhanced inflammatory responses driven by CHIKV. These knockout mice showed increased neutrophil accumulation and augmented tissue damage at the site of infection compared with control mice. Conversely, treatment of wild-type animals with the AnxA1 mimetic peptide (Ac2–26) reduced neutrophil accumulation, decreased local concentration of inflammatory mediators and diminished mechanical hypernociception and paw edema induced by CHIKV-infection. Alterations in viral load were mild both in genetic deletion or with treatment. Combined, our data suggest that the AnxA1-FPR2/ALX pathway is a potential therapeutic strategy to control CHIKV-induced acute inflammation and polyarthralgia.
Collapse
Affiliation(s)
- Simone de Araújo
- Graduate Program in Biological Sciences Physiology and Pharmacology, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Victor R. de Melo Costa
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Franciele M. Santos
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Carla D. Ferreira de Sousa
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Thaiane P. Moreira
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Matheus R. Gonçalves
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Franciel B. Félix
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Celso M. Queiroz-Junior
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Gabriel H. Campolina-Silva
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Department of Obstetrics, Gynecology and Reproduction, CHU de Quebec Research Center (CHUL), Université Laval, Quebec, QC G1V 0A6, Canada
| | - Maurício Lacerda Nogueira
- Department of Dermatological, Infections, and Parasitic Diseases, School of Medicine (FAMERP), São José do Rio Preto, São Paulo 15090-000, Brazil
| | - Michelle A. Sugimoto
- Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK
| | - Caio S. Bonilha
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Center for Research on Inflammatory Diseases, University of São Paulo, São Paulo 05508-000, Brazil
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8QQ, UK
| | - Mauro Perretti
- Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London E1 4NS, UK
| | - Danielle G. Souza
- Graduate Program in Microbiology, Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Vivian V. Costa
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Graduate Program in Cell Biology, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Correspondence: (V.V.C.); (M.M.T.); Tel.: +55-31-3409-3082 (V.V.C.); +55-31-3409-2651 (M.M.T.)
| | - Mauro M. Teixeira
- Drug Research and Development Center, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Correspondence: (V.V.C.); (M.M.T.); Tel.: +55-31-3409-3082 (V.V.C.); +55-31-3409-2651 (M.M.T.)
| |
Collapse
|
33
|
Traverse EM, Millsapps EM, Underwood EC, Hopkins HK, Young M, Barr KL. Chikungunya Immunopathology as It Presents in Different Organ Systems. Viruses 2022; 14:v14081786. [PMID: 36016408 PMCID: PMC9414582 DOI: 10.3390/v14081786] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 12/13/2022] Open
Abstract
Chikungunya virus (CHIKV) is currently an urgent public health problem as high morbidity from the virus leaves populations with negative physical, social, and economic impacts. CHIKV has the potential to affect every organ of an individual, leaving patients with lifelong impairments which negatively affect their quality of life. In this review, we show the importance of CHIKV in research and public health by demonstrating the immunopathology of CHIKV as it presents in different organ systems. Papers used in this review were found on PubMed, using “chikungunya and [relevant organ system]”. There is a significant inflammatory response during CHIKV infection which affects several organ systems, such as the brain, heart, lungs, kidneys, skin, and joints, and the immune response to CHIKV in each organ system is unique. Whilst there is clinical evidence to suggest that serious complications can occur, there is ultimately a lack of understanding of how CHIKV can affect different organ systems. It is important for clinicians to understand the risks to their patients.
Collapse
|
34
|
Sandenon Seteyen AL, Girard-Valenciennes E, Septembre-Malaterre A, Gasque P, Guiraud P, Sélambarom J. Anti-Alphaviral Alkaloids: Focus on Some Isoquinolines, Indoles and Quinolizidines. Molecules 2022; 27:molecules27165080. [PMID: 36014321 PMCID: PMC9416297 DOI: 10.3390/molecules27165080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
The discovery and the development of safe and efficient therapeutics against arthritogenic alphaviruses (e.g., chikungunya virus) remain a continuous challenge. Alkaloids are structurally diverse and naturally occurring compounds in plants, with a wide range of biological activities including beneficial effects against prominent pathogenic viruses and inflammation. In this short review, we discuss the effects of some alkaloids of three biologically relevant structural classes (isoquinolines, indoles and quinolizidines). Based on various experimental models (viral infections and chronic diseases), we highlight the immunomodulatory effects of these alkaloids. The data established the capacity of these alkaloids to interfere in host antiviral and inflammatory responses through key components (antiviral interferon response, ROS production, inflammatory signaling pathways and pro- and anti-inflammatory cytokines production) also involved in alphavirus infection and resulting inflammation. Thus, these data may provide a convincing perspective of research for the use of alkaloids as immunomodulators against arthritogenic alphavirus infection and induced inflammation.
Collapse
Affiliation(s)
- Anne-Laure Sandenon Seteyen
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France
| | - Emmanuelle Girard-Valenciennes
- Laboratoire de Chimie et de Biotechnologie des Produits Naturels (CHEMBIOPRO), Université de La Réunion, 97400 Saint-Denis, France
| | - Axelle Septembre-Malaterre
- Centre Hospitalier Universitaire de La Réunion, Laboratoire d’Immunologie Clinique et Expérimentale de la Zone Océan Indien (LICE-OI), Pôle de Biologie, 97400 Saint-Denis, France
| | - Philippe Gasque
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France
- Centre Hospitalier Universitaire de La Réunion, Laboratoire d’Immunologie Clinique et Expérimentale de la Zone Océan Indien (LICE-OI), Pôle de Biologie, 97400 Saint-Denis, France
| | - Pascale Guiraud
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France
| | - Jimmy Sélambarom
- Unité de Recherche Etudes Pharmaco-Immunologiques (UR-EPI), Université de La Réunion, 97400 Saint-Denis, France
- Correspondence:
| |
Collapse
|
35
|
Valdés-López JF, Velilla P, Urcuqui-Inchima S. Vitamin D modulates the expression of Toll-like receptors and pro-inflammatory cytokines without affecting Chikungunya virus replication, in monocytes and macrophages. Acta Trop 2022; 232:106497. [PMID: 35508271 DOI: 10.1016/j.actatropica.2022.106497] [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: 03/11/2022] [Revised: 04/12/2022] [Accepted: 04/30/2022] [Indexed: 11/30/2022]
Abstract
Chikungunya virus (CHIKV) is a zoonotic arthropod-borne virus that causes Chikungunya fever (CHIKF), a self-limiting disease characterized by myalgia and acute or chronic arthralgia. CHIKF pathogenesis has an important immunological component since higher levels of pro-inflammatory factors, including cytokines and chemokines, are detected in CHIKV-infected patients. In vitro studies, using monocytes and macrophages have shown that CHIKV infection promotes elevated production of pro-inflammatory cytokines and antiviral response factors. Vitamin D3 (VD3) has been described as an important modulator of immune response and as an antiviral factor for several viruses. Here, we aimed to study the effects of VD3 treatment on viral replication and pro-inflammatory response in CHIKV-infected human monocytes (VD3-Mon) and monocyte-derived macrophages differentiated in the absence (MDMs) or the presence of VD3 (VD3-MDMs). We found that VD3 treatment did not suppress CHIKV replication in either VD3-Mon or VD3-MDMs. However, the expression of VDR, CAMP and CYP24A1 mRNAs was altered by CHIKV infection. Furthermore, VD3 treatment alters TLRs mRNA expression and production of pro-inflammatory cytokines, including TNFα and CXCL8/IL8, but not IL1β and IL6, in response to CHIKV infection in both VD3-Mon and VD3-MDMs. While a significant decrease in CXCL8/IL8 production was observed in CHIKV-infected VD3-Mon, significantly higher production of CXCL8/IL8 was observed in CHIKV-infected VD3-MDM at 24 hpi. Altogether, our results suggest that vitamin D3 may play an important role in ameliorating pro-inflammatory response during CHIKV infection in human Mon, but not in MDMs. Although further studies are needed to evaluate the efficacy of VD3; nevertheless, this study provides novel insights into its benefits in modulating the inflammatory response elicited by CHIKV infection in humans.
Collapse
Affiliation(s)
- Juan Felipe Valdés-López
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| | - Paula Velilla
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
36
|
Restrepo BN, Marín K, Romero P, Arboleda M, Muñoz AL, Bosch I, Vásquez-Serna H, Torres OA. Role of cytokines, chemokines, C3a, and mannose-binding lectin in the evolution of the chikungunya infection. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2022; 11:51-63. [PMID: 35874468 PMCID: PMC9301056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 06/12/2022] [Indexed: 06/15/2023]
Abstract
UNLABELLED The pathogenesis of the severity of chikungunya infection is not yet fully understood. OBJECTIVE To assess the role of the cytokines/chemokines and system of complement in the evolution of chikungunya infection. METHODS In both acute and chronic phases, we measured the serum levels of 12 cytokines/chemokines and two complement mediators: mannose-binding lectin (MBL) and C3a, in 83 patients with chikungunya infection and ten healthy controls. RESULTS During the acute phase, 75.9% of the patients developed musculoskeletal disorders, and in 37.7% of them, these disorders persisted until the chronic phase. In general, patients had higher levels of cytokines than healthy controls, with significant differences for IFN-γ, IL-6, IL-8, IL-10, and MIP-1. Most cytokines exhibited a downward trend during the chronic phase. However, only IL-10, and MIP-1 levels were significantly lower in the chronic phase. Additionally, these levels never decreased to concentrations found in healthy controls. Moreover, MBL levels were significantly higher in the acute phase compared with the chronic phase. C3a levels were significantly higher in patients with musculoskeletal disorder compared with patients without it, in both acute-phase 118.2 (66.5-252.9), and chronic phase 68.5 (64.4-71.3), P < 0.001. Interestingly, C3a levels were significantly higher when patients had a severe disease version. Besides, in the acute phase, C3a levels were higher in patients that suffer arthritis as opposed to when they suffer arthralgia, 194.3 (69.5-282.2), and 70.9 (62.4-198.8), P = 0.013, respectively. CONCLUSIONS Our results showed an immunological response that persisted until the chronic phase and the role of the complement system in the severity of the disease.
Collapse
Affiliation(s)
- Berta N Restrepo
- Instituto Colombiano de Medicina Tropical, Universidad CESSabaneta, Antioquía, Colombia
| | - Katerine Marín
- Instituto Colombiano de Medicina Tropical, Universidad CESSabaneta, Antioquía, Colombia
| | - Paola Romero
- Instituto Colombiano de Medicina Tropical, Universidad CESSabaneta, Antioquía, Colombia
| | - Margarita Arboleda
- Instituto Colombiano de Medicina Tropical, Universidad CESSabaneta, Antioquía, Colombia
| | - Ana L Muñoz
- Science Faculty, Universidad Antonio NariñoBogotá, Cundinamarca, Colombia
| | - Irene Bosch
- Institute for Medical Engineering & Science, Massachusetts Institute of TechnologyCambridge, United States
| | | | - Orlando A Torres
- Faculty of Veterinary Medicine, Universidad Antonio NariñoBogotá, Cundinamarca, Colombia
| |
Collapse
|
37
|
Roques P, Fritzer A, Dereuddre-Bosquet N, Wressnigg N, Hochreiter R, Bossevot L, Pascal Q, Guehenneux F, Bitzer A, Corbic Ramljak I, Le Grand R, Lundberg U, Meinke A. Effectiveness of CHIKV vaccine VLA1553 demonstrated by passive transfer of human sera. JCI Insight 2022; 7:160173. [PMID: 35700051 PMCID: PMC9431671 DOI: 10.1172/jci.insight.160173] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
Chikungunya virus (CHIKV) is a reemerging mosquito-borne alphavirus responsible for numerous outbreaks. Chikungunya can cause debilitating acute and chronic disease. Thus, the development of a safe and effective CHIKV vaccine is an urgent global health priority. This study evaluated the effectiveness of the live-attenuated CHIKV vaccine VLA1553 against WT CHIKV infection by using passive transfer of sera from vaccinated volunteers to nonhuman primates (NHP) subsequently exposed to WT CHIKV and established a serological surrogate of protection. We demonstrated that human VLA1553 sera transferred to NHPs conferred complete protection from CHIKV viremia and fever after challenge with homologous WT CHIKV. In addition, serum transfer protected animals from other CHIKV-associated clinical symptoms and from CHIKV persistence in tissue. Based on this passive transfer study, a 50% micro–plaque reduction neutralization test titer of ≥ 150 was determined as a surrogate of protection, which was supported by analysis of samples from a seroepidemiological study. In conclusion, considering the unfeasibility of an efficacy trial due to the unpredictability and explosive, rapidly moving nature of chikungunya outbreaks, the definition of a surrogate of protection for VLA1553 is an important step toward vaccine licensure to reduce the medical burden caused by chikungunya.
Collapse
Affiliation(s)
- Pierre Roques
- Unité de Virologie, Commissariat à l'énergie atomique et aux énergies alternatives, Fontenay-aux-Roses, France
| | | | | | - Nina Wressnigg
- Clinical Strategy, Valneva Austria GmbH, Vienna, Austria
| | | | - Laetitia Bossevot
- DSV/IMETI, Commissariat à l'énergie atomique et aux énergies alternatives, Fontenay-aux-Roses, France
| | - Quentin Pascal
- DSV/IMETI, Commissariat à l'énergie atomique et aux énergies alternatives, Fontenay-aux-Roses, France
| | | | | | | | - Roger Le Grand
- DSV/IMETI, Commissariat à l'énergie atomique et aux énergies alternatives, Fontenay-aux-Roses, France
| | | | | |
Collapse
|
38
|
A Review on Chikungunya Virus Epidemiology, Pathogenesis and Current Vaccine Development. Viruses 2022; 14:v14050969. [PMID: 35632709 PMCID: PMC9147731 DOI: 10.3390/v14050969] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 12/20/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that recently re-emerged in many parts of the world causing large-scale outbreaks. CHIKV infection presents as a febrile illness known as chikungunya fever (CHIKF). Infection is self-limited and characterized mainly by severe joint pain and myalgia that can last for weeks or months; however, severe disease presentation can also occur in a minor proportion of infections. Among the atypical CHIKV manifestations that have been described, severe arthralgia and neurological complications, such as encephalitis, meningitis, and Guillain–Barré Syndrome, are now reported in many outbreaks. Moreover, death cases were also reported, placing CHIKV as a relevant public health disease. Virus evolution, globalization, and climate change may have contributed to CHIKV spread. In addition to this, the lack of preventive vaccines and approved antiviral treatments is turning CHIKV into a major global health threat. In this review, we discuss the current knowledge about CHIKV pathogenesis, with a focus on atypical disease manifestations, such as persistent arthralgia and neurologic disease presentation. We also bring an up-to-date review of the current CHIKV vaccine development. Altogether, these topics highlight some of the most recent advances in our understanding of CHIKV pathogenesis and also provide important insights into the current development and clinical trials of CHIKV potential vaccine candidates.
Collapse
|
39
|
Increased Indoleamine 2,3-Dioxygenase 1 (IDO-1) Activity and Inflammatory Responses during Chikungunya Virus Infection. Pathogens 2022; 11:pathogens11040444. [PMID: 35456119 PMCID: PMC9028473 DOI: 10.3390/pathogens11040444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 12/13/2022] Open
Abstract
Chikungunya virus (CHIKV) infection causes intense cytokine/chemokine inflammatory responses and debilitating joint pain. Indoleamine2,3–dioxygenase 1 (IDO-1) is an enzyme that initiates the tryptophan degradation that is important in initial host innate immune defense against infectious pathogens. Besides that, IDO-1 activation acts as a regulatory mechanism to prevent overactive host immune responses. In this study, we evaluated IDO-1 activity and cytokine/chemokine patterns in CHIKV patients. Higher IDO-1 (Kyn/Trp ratio) activation was observed during the early acute phase of CHIKV infection and declined in the chronic phase. Importantly, increased concentrations of Tumor Necrosis Factor-α (TNF-α), Interleukin-6 (IL-6), Interferon γ (IFN-γ), C-C motif chemokine ligand 2/Monocyte Chemoattractant Protein-1 (CCL2/MCP-1) and C-X-C motif chemokine ligand 10/Interferon Protein-10 (CXCL10/IP-10) were found in the acute phase of infection, while C-C motif chemokine ligand 4/Macrophage Inflammatory Protein 1 β (CCL4/MIP-1β) was found at increased concentrations in the chronic phase. Likewise, CHIKV patients with arthritis had significantly higher concentrations of CCL4/MIP-1β compared to patients without arthritis. Taken together, these data demonstrated increased IDO-1 activity, possibly exerting both antiviral effects and regulating exacerbated inflammatory responses. CCL4/MIP-1β may have an important role in the persistent inflammation and arthritic symptoms following chikungunya infection.
Collapse
|
40
|
Abstract
CHIKV has been prevalent in Africa, Asia, and the Indian Ocean Islands for decades. There are currently no clinically approved vaccines or specific antiviral drugs targeting CHIKV.
Collapse
|
41
|
Locke MC, Fox LE, Dunlap BF, Young AR, Monte K, Lenschow DJ. Interferon Alpha, but Not Interferon Beta, Acts Early To Control Chronic Chikungunya Virus Pathogenesis. J Virol 2022; 96:e0114321. [PMID: 34668781 PMCID: PMC8754211 DOI: 10.1128/jvi.01143-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/11/2021] [Indexed: 11/20/2022] Open
Abstract
Chikungunya virus (CHIKV) is an arthritogenic alphavirus that causes both debilitating acute and chronic disease. Previous work has shown that type I interferons (IFNs) play a critical role in limiting CHIKV pathogenesis and that interferon alpha (IFN-α) and interferon beta (IFN-β) control acute CHIKV infection by distinct mechanisms. However, the role of type I IFNs, especially specific subtypes, during chronic CHIKV disease is unclear. To address this gap in knowledge, we evaluated chronic CHIKV pathogenesis in mice lacking IFN-α or IFN-β. We found that IFN-α was the dominant subtype that controls chronic disease. Despite detecting a varying type I IFN response throughout the course of disease, IFN-α acts within the first few days of infection to control the levels of persistent CHIKV RNA. In addition, using a novel CHIKV-3'-Cre tdTomato reporter system that fate maps CHIKV-infected cells, we showed that IFN-α limits the number of cells that survive CHIKV at sites of dissemination, particularly dermal fibroblasts and immune cells. Though myofibers play a significant role in CHIKV disease, they were not impacted by the loss of IFN-α. Our studies highlight that IFN-α and IFN-β play divergent roles during chronic CHIKV disease through events that occur early in infection and that not all cell types are equally dependent on type I IFNs for restricting viral persistence. IMPORTANCE Chikungunya virus (CHIKV) is a reemerging global pathogen with no effective vaccine or antiviral treatment for acute or chronic disease, and the mechanisms underlying chronic disease manifestations remain poorly defined. The significance of our research is in defining IFN-α, but not IFN-β, as an important host regulator of chronic CHIKV pathogenesis that acts within the first 48 hours of infection to limit persistent viral RNA and the number of cells that survive CHIKV infection 1 month post-infection. Loss of IFN-α had a greater impact on immune cells and dermal fibroblasts than myofibers, highlighting the need to delineate cell-specific responses to type I IFNs. Altogether, our work demonstrates that very early events of acute CHIKV infection influence chronic disease. Continued efforts to delineate early host-pathogen interactions may help stratify patients who are at risk for developing chronic CHIKV symptoms and identify therapeutics that may prevent progression to chronic disease altogether.
Collapse
Affiliation(s)
- Marissa C. Locke
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Lindsey E. Fox
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Bria F. Dunlap
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Alissa R. Young
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Kristen Monte
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Deborah J. Lenschow
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
42
|
Rocha DCP, Souza TMA, Nunes PCG, Mohana-Borges R, Paes MV, Guimarães GMC, Arcila JCS, Paiva IA, Azeredo ELD, Damasco PV, de Souza LJ, Dos Santos FB, Allonso D. Increased circulating levels of High Mobility Group Box 1 (HMGB1) in acute-phase Chikungunya virus infection: Potential disease biomarker. J Clin Virol 2021; 146:105054. [PMID: 34920373 DOI: 10.1016/j.jcv.2021.105054] [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: 04/13/2021] [Revised: 11/01/2021] [Accepted: 12/08/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Chikungunya virus (CHIKV) causes a febrile syndrome with intense and debilitating arthralgia that can persist for several months or years after complete virus clearance. As there is no specific antiviral treatment or vaccine against CHIKV, identification of serological markers that help clinical management of CHIKV patients is urgent. The High Mobility Group Box 1 (HMGB1) protein is secreted to extracellular milieu and triggers an intense inflammatory process by inducing the overexpression of pro-inflammatory cytokines. HMGB1 plays an important role in several virus diseases as well as in rheumatoid arthritis. OBJECTIVES This study focus on the investigation of HMGB1 serum levels in a sera panel from CHIKV-infected patients in an attempt to assess its potential as a biomarker for chikungunya clinical management. STUDY DESIGN Eighty CHIKV-positive samples and 32 samples from healthy donors were subjected to a quantitative HMGB1 ELISA assay to assess the HMGB1 circulating levels. RESULTS HMGB1 levels were significantly higher in CHIKV-positive samples (516.12 ng/mL, SEM ± 48.83 ng/mL) compared to negative control (31.20 ng/mL, SEM ± 3.24 ng/mL, p < 0.0001). Circulating levels of HMGB1 persisted elevated during the whole acute-phase of disease and correlated with virus titer (p < 0.05). CONCLUSIONS The present study is the first to describe increased serum levels of HMGB1 in CHIKV infection and its positive correlation with virus titer, suggesting its potential use as a biomarker for diagnosis and treatment of chikungunya fever.
Collapse
Affiliation(s)
- Daniele C P Rocha
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil; Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - Thiara Manuelle Alves Souza
- Laboratório de Imunologia Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ 21040-900, Brazil
| | - Priscila Conrado Guerra Nunes
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer, Rio de Janeiro, RJ 20231-092, Brazil; Superintendência de Informações Estratégicas de Vigilância em Saúde (SIEVS/RJ), Secretaria de Saúde, Governo do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20031-142, Brazil
| | - Ronaldo Mohana-Borges
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - Marciano V Paes
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, RJ 21040-900, Brazil
| | - Gabriel M C Guimarães
- Laboratório de Imunologia Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ 21040-900, Brazil
| | - Juan C S Arcila
- Laboratório de Imunologia Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ 21040-900, Brazil
| | - Iury Amâncio Paiva
- Laboratório de Imunologia Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ 21040-900, Brazil
| | - Elzinandes Leal de Azeredo
- Laboratório de Imunologia Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ 21040-900, Brazil
| | - Paulo Vieira Damasco
- Hospital Universitário Gaffrée Guinle, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro, RJ 20270-004, Brazil; Hospital Universitário Pedro Ernesto, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, RJ 20551-030, Brazil
| | - Luiz José de Souza
- Hospital dos Plantadores de Cana, Campos dos Goytacazes, RJ 28025-496, Brazil
| | - Flavia B Dos Santos
- Laboratório de Imunologia Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ 21040-900, Brazil
| | - Diego Allonso
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| |
Collapse
|
43
|
Chikungunya Virus Exposure Partially Cross-Protects against Mayaro Virus Infection in Mice. J Virol 2021; 95:e0112221. [PMID: 34549980 DOI: 10.1128/jvi.01122-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Chikungunya virus (CHIKV) and Mayaro virus (MAYV) are closely related members of the Semliki Forest virus antigenic complex classified as belonging to the genus Alphavirus of the family Togaviridae. These viruses cause human disease, with sudden fever and joint inflammation that can persist for long periods. CHIKV is the causative agent of large outbreaks worldwide, and MAYV infection represents a growing public health concern in Latin America, causing sporadic cases and geographically limited outbreaks. Considering the relationship between CHIKV and MAYV, the present study aimed to evaluate if preexisting CHIKV immunity protects against MAYV infection. Immunocompetent C57BL/6 mice were intraperitoneally infected with CHIKV and, 4 weeks later, they were infected with MAYV in their hind paw. We observed that the preexistence of CHIKV immunity conferred partial cross-protection against secondary MAYV infection, reducing disease severity, tissue viral load, and histopathological scores. Interestingly, CHIKV antibodies from humans and mice showed low cross-neutralization to MAYV, but neutralizing activity significantly increased after secondary infection. Furthermore, depletion of adaptive immune cells (CD4+ T, CD8+ T, and CD19+ B cells) did not alter the cross-protection phenotype, suggesting that distinct cell subsets or a combination of adaptive immune cells stimulated by CHIKV are responsible for the partial cross-protection against MAYV. The reduction of proinflammatory cytokines, such as interferon gamma (IFN-γ), in animals secondarily infected by MAYV, suggests a role for innate immunity in cross-protection. Our findings shed light on how preexisting immunity to arthritogenic alphaviruses may affect secondary infection, which may further develop relevant influence in disease outcome and viral transmission. IMPORTANCE Mosquito-borne viruses have a worldwide impact, especially in tropical climates. Chikungunya virus has been present mostly in developing countries, causing millions of infections, while Mayaro virus, a close relative, has been limited to the Caribbean and tropical regions of Latin America. The potential emergence and spread of Mayaro virus to other high-risk areas have increased the scientific community's attention to an imminent worldwide epidemic. Here, we designed an experimental protocol of chikungunya and Mayaro virus mouse infection, which develops a measurable and quantifiable disease that allows us to make inferences about potential immunological effects during secondary virus infection. Our results demonstrate that previous chikungunya virus infection is able to reduce the severity of clinical outcomes during secondary Mayaro infection. We provide scientific understanding of immunological features during secondary infection with the closely related virus, thus assisting in better comprehending viral transmission and the pathological outcome of these diseases.
Collapse
|
44
|
Constant LEC, Rajsfus BF, Carneiro PH, Sisnande T, Mohana-Borges R, Allonso D. Overview on Chikungunya Virus Infection: From Epidemiology to State-of-the-Art Experimental Models. Front Microbiol 2021; 12:744164. [PMID: 34675908 PMCID: PMC8524093 DOI: 10.3389/fmicb.2021.744164] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/07/2021] [Indexed: 12/27/2022] Open
Abstract
Chikungunya virus (CHIKV) is currently one of the most relevant arboviruses to public health. It is a member of the Togaviridae family and alphavirus genus and causes an arthritogenic disease known as chikungunya fever (CHIKF). It is characterized by a multifaceted disease, which is distinguished from other arbovirus infections by the intense and debilitating arthralgia that can last for months or years in some individuals. Despite the great social and economic burden caused by CHIKV infection, there is no vaccine or specific antiviral drugs currently available. Recent outbreaks have shown a change in the severity profile of the disease in which atypical and severe manifestation lead to hundreds of deaths, reinforcing the necessity to understand the replication and pathogenesis processes. CHIKF is a complex disease resultant from the infection of a plethora of cell types. Although there are several in vivo models for studying CHIKV infection, none of them reproduces integrally the disease signature observed in humans, which is a challenge for vaccine and drug development. Therefore, understanding the potentials and limitations of the state-of-the-art experimental models is imperative to advance in the field. In this context, the present review outlines the present knowledge on CHIKV epidemiology, replication, pathogenesis, and immunity and also brings a critical perspective on the current in vitro and in vivo state-of-the-art experimental models of CHIKF.
Collapse
Affiliation(s)
- Larissa E. C. Constant
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Bia F. Rajsfus
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro H. Carneiro
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tháyna Sisnande
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ronaldo Mohana-Borges
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diego Allonso
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
45
|
Gupta S, Mishra KP, Gupta R, Singh SB. Andrographolide - A prospective remedy for chikungunya fever and viral arthritis. Int Immunopharmacol 2021; 99:108045. [PMID: 34435582 DOI: 10.1016/j.intimp.2021.108045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 08/01/2021] [Accepted: 08/01/2021] [Indexed: 12/14/2022]
Abstract
AIM Andrographolide, the major bioactive compound of the plant Andrographis paniculata, exerts anti-inflammatory, cyto-, neuro- and hepato-protective effects. Traditional remedies for infectious diseases include A. paniculata for maladies like fever, pain, rashes which are associated with chikungunya and other arboviral diseases. Since andrographolide and A. paniculata have potent antiviral properties, the present review aims to provide a comprehensive report of symptoms and immunological molecules involved in chikungunya virus (CHIKV) infection and the therapeutic role of andrographolide in the mitigation of chikungunya and associated symptoms. MATERIALS AND METHODS Studies on the therapeutic role of A. paniculata and andrographolide in chikungunya and other viral infections published between 1991 and 2021 were searched on various databases. RESULTS AND DISCUSSION The havoc created by chikungunya is due to the associated debilitating symptoms including arthralgia and myalgia which sometimes remains for years. The authors reviewed and summarized the various symptoms and immunological molecules related to CHIKV replication and associated inflammation, oxidative and unfolded protein stress, apoptosis and arthritis. Additionally, the authors suggested andrographolide as a remedy for chikungunya and other arboviral infections by highlighting its role in the regulation of molecules involved in unfolded protein response pathway, immunomodulation, inflammation, virus multiplication, oxidative stress, apoptosis and arthritis. CONCLUSION The present review demonstrated the major complications associated with chikungunya and the role of andrographolide in alleviating the chikungunya associated symptoms to encourage further investigations using this promising compound towards early development of an anti-CHIKV drug. Chemical Compound studied: andrographolide (PubChem CID: 5318517).
Collapse
Affiliation(s)
- Swati Gupta
- Division of Epidemiology and Communicable Diseases, Indian Council of Medical Research (ICMR), Ansari Nagar, New Delhi 110029, India.
| | - K P Mishra
- Defence Research and Development Organization (DRDO)-HQ, Rajaji Marg, New Delhi 110011, India
| | - Rupali Gupta
- Department of Neurology, Duke University Medical Center, Durham, NC, United States
| | - S B Singh
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
46
|
Queiroz JL, Maciel MSC, Teles LDS, Vieira SMA, Gomes TN, Fernandes HF, de Oliveira JS, Ferreira GP, Pereira ACTDC. Prevalence and Influence of the -174 G/C Polymorphism in the Interleukin-6 Gene in Arboviruses Infections. Viral Immunol 2021; 34:559-566. [PMID: 34415813 DOI: 10.1089/vim.2020.0256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Dengue virus and Chikungunya virus are arboviruses that affect thousands of people worldwide annually. The mechanisms involved in viral pathogenesis still need to be better understood. Single nucleotide polymorphisms (SNPs) in immune genes may be involved in the protection, susceptibility, and/or progression of these diseases. This study was performed to investigate the SNP -174 G/C in the interleukin-6 (IL-6) gene in patients with dengue or chikungunya from Northeastern Brazil. A total of 581 blood samples were analyzed, of which 244 were part of the negative control group, genomic DNA was extracted, and the SNP was genotyped using real-time polymerase chain reaction (PCR). The data obtained were used to conduct statistical analyses of the genotype and allele frequencies. We suggest that the G/C genotype and C allele of the SNP -174 G/C in the IL-6 gene are related to protection against dengue in the studied population. No significant differences were observed in chikungunya patients. This is the first study that assessed the association of the SNP -174 G/C in patients with chikungunya. We identified the presence of the C allele as a protective factor against dengue in the studied population.
Collapse
Affiliation(s)
- Julianna Lima Queiroz
- Programa de Pós-Graduação em Ciências Biomédicas, and Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil.,Laboratório de Biologia de Microrganismos, Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil
| | - Mariella Sousa Coelho Maciel
- Laboratório de Biologia de Microrganismos, Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil.,Graduação em Biomedicina, Universidade Federal do Piauí, Parnaíba, Brazil
| | - Lorena da Silva Teles
- Laboratório de Biologia de Microrganismos, Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil.,Graduação em Biomedicina, Universidade Federal do Piauí, Parnaíba, Brazil
| | - Silveny Meiga Alves Vieira
- Programa de Pós-Graduação em Ciências Biomédicas, and Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil.,Laboratório de Biologia de Microrganismos, Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil
| | - Thiago Nobre Gomes
- Programa de Pós-Graduação em Ciências Biomédicas, and Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil.,Laboratório de Biologia de Microrganismos, Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil
| | | | - Jefferson Soares de Oliveira
- Programa de Pós-Graduação em Ciências Biomédicas, and Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil.,Laboratório de Biologia de Microrganismos, Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil
| | - Gustavo Portela Ferreira
- Programa de Pós-Graduação em Ciências Biomédicas, and Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil.,Laboratório de Biologia de Microrganismos, Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil
| | - Anna Carolina Toledo da Cunha Pereira
- Programa de Pós-Graduação em Ciências Biomédicas, and Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil.,Laboratório de Biologia de Microrganismos, Universidade Federal do Delta do Parnaíba, Parnaíba, Brazil
| |
Collapse
|
47
|
Kril V, Aïqui-Reboul-Paviet O, Briant L, Amara A. New Insights into Chikungunya Virus Infection and Pathogenesis. Annu Rev Virol 2021; 8:327-347. [PMID: 34255544 DOI: 10.1146/annurev-virology-091919-102021] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chikungunya virus (CHIKV) is a re-emerging mosquito-borne alphavirus responsible for major outbreaks of disease since 2004 in the Indian Ocean islands, South east Asia, and the Americas. CHIKV causes debilitating musculoskeletal disorders in humans that are characterized by fever, rash, polyarthralgia, and myalgia. The disease is often self-limiting and nonlethal; however, some patients experience atypical or severe clinical manifestations, as well as a chronic rheumatic syndrome. Unfortunately, no efficient antivirals against CHIKV infection are available so far, highlighting the importance of deepening our knowledge of CHIKV host cell interactions and viral replication strategies. In this review, we discuss recent breakthroughs in the molecular mechanisms that regulate CHIKV infection and lay down the foundations to understand viral pathogenesis. We describe the role of the recently identified host factors co-opted by the virus for infection and pathogenesis, and emphasize the importance of CHIKV nonstructural proteins in both replication complex assembly and host immune response evasion. Expected final online publication date for the Annual Review of Virology, Volume 8 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Vasiliya Kril
- Biology of Emerging Virus Team, INSERM U944, CNRS UMR 7212, Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, 75010 Paris, France;
| | - Olivier Aïqui-Reboul-Paviet
- RNA Viruses and Metabolism Team, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, University of Montpellier, 34293 Montpellier, France;
| | - Laurence Briant
- RNA Viruses and Metabolism Team, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, University of Montpellier, 34293 Montpellier, France;
| | - Ali Amara
- Biology of Emerging Virus Team, INSERM U944, CNRS UMR 7212, Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, 75010 Paris, France;
| |
Collapse
|
48
|
Dhenni R, Yohan B, Alisjahbana B, Lucanus A, Riswari SF, Megawati D, Haryanto S, Gampamole D, Hayati RF, Sari K, Witari NPD, Myint KSA, Sasmono RT. Comparative cytokine profiling identifies common and unique serum cytokine responses in acute chikungunya and dengue virus infection. BMC Infect Dis 2021; 21:639. [PMID: 34215212 PMCID: PMC8254284 DOI: 10.1186/s12879-021-06339-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 06/18/2021] [Indexed: 01/09/2023] Open
Abstract
Background Infection by chikungunya (CHIKV) and dengue virus (DENV) can cause a wide spectrum of clinical features, many of which are undifferentiated. Cytokines, which broadly also include chemokines and growth factors, have been shown to play a role in protective immunity as well as DENV and CHIKV pathogenesis. However, differences in cytokine response to both viruses remain poorly understood, especially in patients from countries where both viruses are endemic. Our study is therefore aimed to provide a comparative profiling of cytokine response induced by acute DENV and CHIKV infections in patients with similar disease stages and in experimental in vitro infections. Methods By using multiplex immunoassay, we compared host cytokine profiles between acute CHIKV and DENV infections by analysing serum cytokine levels of IL-1α, IL-4, IL-5, IL-8, IL-13, RANTES, MCP-3, eotaxin, PDGF-AB/BB, and FGF-2 from the sera of acute chikungunya and dengue fever patients. We further investigated the cytokine profile responses using experimental in vitro CHIKV and DENV infections of peripheral blood mononuclear cells (PBMCs). Results We found that both CHIKV and DENV-infected patients had an upregulated level of IL-8 and IL-4, with the highest IL-4 level observed in DENV-2 infected patients. Higher IL-8 level was also correlated with lower platelet count in dengue patients. IL-13 and MCP-3 downregulation was observed only in chikungunya patients, while conversely PDGF-AB/BB and FGF-2 downregulation was unique in dengue patients. Age-associated differential expression of IL-13, MCP-3, and IL-5 was also observed, while distinct kinetics of IL-4, IL-8, and FGF-2 expression between CHIKV and DENV-infected patients were identified. Furthermore, the unique pattern of IL-8, IL-13 and MCP-3, but not IL-4 expression was also recapitulated using experimental in vitro infection in PBMCs. Conclusions Taken together, our study identified common cytokine response profile characterized by upregulation of IL-8 and IL-4 between CHIKV and DENV infection. Downregulation of IL-13 and MCP-3 was identified as a unique cytokine response profile of acute CHIKV infection, while distinct downregulation of PDGF-AB/BB and FGF-2 characterized the response from acute DENV infection. Our study provides an important overview of the host cytokine responses between CHIKV and DENV infection, which is important to further understand the mechanism and pathology of these diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06339-6.
Collapse
Affiliation(s)
- Rama Dhenni
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | | | | | - Anton Lucanus
- School of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Australia
| | | | - Dewi Megawati
- Faculty of Medicine and Health Sciences, Warmadewa University, Denpasar, Bali, Indonesia
| | | | | | - Rahma F Hayati
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | - Kartika Sari
- Faculty of Medicine and Health Sciences, Warmadewa University, Denpasar, Bali, Indonesia
| | - Ni Putu Diah Witari
- Faculty of Medicine and Health Sciences, Warmadewa University, Denpasar, Bali, Indonesia
| | | | - R Tedjo Sasmono
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia.
| |
Collapse
|
49
|
Hibl BM, Dailey Garnes NJM, Kneubehl AR, Vogt MB, Spencer Clinton JL, Rico-Hesse RR. Mosquito-bite infection of humanized mice with chikungunya virus produces systemic disease with long-term effects. PLoS Negl Trop Dis 2021; 15:e0009427. [PMID: 34106915 PMCID: PMC8189471 DOI: 10.1371/journal.pntd.0009427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/02/2021] [Indexed: 12/13/2022] Open
Abstract
Chikungunya virus (CHIKV) is an emerging, mosquito-borne alphavirus responsible for acute to chronic arthralgias and neuropathies. Although it originated in central Africa, recent reports of disease have come from many parts of the world, including the Americas. While limiting human CHIKV cases through mosquito control has been used, it has not been entirely successful. There are currently no licensed vaccines or treatments specific for CHIKV disease, thus more work is needed to develop effective countermeasures. Current animal research on CHIKV is often not representative of human disease. Most models use CHIKV needle inoculation via unnatural routes to create immediate viremia and localized clinical signs; these methods neglect the natural route of transmission (the mosquito vector bite) and the associated human immune response. Since mosquito saliva has been shown to have a profound effect on viral pathogenesis, we evaluated a novel model of infection that included the natural vector, Aedes species mosquitoes, transmitting CHIKV to mice containing components of the human immune system. Humanized mice infected by 3-6 mosquito bites showed signs of systemic infection, with demonstrable viremia (by qRT-PCR and immunofluorescent antibody assay), mild to moderate clinical signs (by observation, histology, and immunohistochemistry), and immune responses consistent with human infection (by flow cytometry and IgM ELISA). This model should give a better understanding of human CHIKV disease and allow for more realistic evaluations of mechanisms of pathogenesis, prophylaxis, and treatments.
Collapse
Affiliation(s)
- Brianne M. Hibl
- Center for Comparative Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Natalie J. M. Dailey Garnes
- Section of Infectious Disease, Department of Internal Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Section of Pediatric Infectious Diseases, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Alexander R. Kneubehl
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Megan B. Vogt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Integrative Molecular and Biomedical Sciences Graduate Program, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jennifer L. Spencer Clinton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Rebecca R. Rico-Hesse
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
50
|
Irinotecan (CPT-11) Canonical Anti-Cancer Drug Can also Modulate Antiviral and Pro-Inflammatory Responses of Primary Human Synovial Fibroblasts. Cells 2021; 10:cells10061431. [PMID: 34201243 PMCID: PMC8230279 DOI: 10.3390/cells10061431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 11/29/2022] Open
Abstract
Alphaviruses are a group of arboviruses that generate chronic inflammatory rheumatisms in humans. Currently, no approved vaccines or antiviral therapies are available to prevent or treat alphavirus-induced diseases. The aim of this study was to evaluate the repositioning of the anti-cancer molecule irinotecan as a potential modulator of the antiviral and inflammatory responses of primary human synovial fibroblasts (HSF), the main stromal cells of the joint synovium. HSF were exposed to O’nyong-nyong virus (ONNV) and polyinosinic-polycytidylic acid (PIC) to mimic, respectively, acute and chronic infectious settings. The cytokine IL-1β was used as a major pro-inflammatory cytokine to stimulate HSF. Quantitative RT-PCR analysis revealed that irinotecan at 15 µM was able to amplify the antiviral response (i.e., interferon-stimulated gene expression) of HSF exposed to PIC and reduce the expression of pro-inflammatory genes (CXCL8, IL-6 and COX-2) upon IL-1β treatment. These results were associated with the regulation of the expression of several genes, including those encoding for STAT1, STAT2, p53 and NF-κB. Irinotecan did not modulate these responses in both untreated cells and cells stimulated with ONNV. This suggests that this drug could be therapeutically useful for the treatment of chronic and severe (rather than acute) arthritis due to viruses.
Collapse
|