Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Vaccine ; 41(26): 3898-3906, 2023 06 13.
Artigo em Inglês | MEDLINE | ID: covidwho-20242800

RESUMO

BACKGROUND: Chikungunya, a mosquito-borne viral disease caused by the chikungunya virus (CHIKV), causes a significant global health burden, and there is currently no approved vaccine to prevent chikungunya disease. In this study, the safety and immunogenicity of a CHIKV mRNA vaccine candidate (mRNA-1388) were evaluated in healthy participants in a CHIKV-nonendemic region. METHODS: This phase 1, first-in-human, randomized, placebo-controlled, dose-ranging study enrolled healthy adults (ages 18-49 years) between July 2017 and March 2019 in the United States. Participants were randomly assigned (3:1) to receive 2 intramuscular injections 28 days apart with mRNA-1388 in 3 dose-level groups (25 µg, 50 µg, and 100 µg) or placebo and were followed for up to 1 year. Safety (unsolicited adverse events [AEs]), tolerability (local and systemic reactogenicity; solicited AEs), and immunogenicity (geometric mean titers [GMTs] of CHIKV neutralizing and binding antibodies) of mRNA-1388 versus placebo were evaluated. RESULTS: Sixty participants were randomized and received ≥ 1 vaccination; 54 (90 %) completed the study. mRNA-1388 demonstrated favorable safety and reactogenicity profiles at all dose levels. Immunization with mRNA-1388 induced substantial and persistent humoral responses. Dose-dependent increases in neutralizing antibody titers were observed; GMTs (95 % confidence intervals [CIs]) at 28 days after dose 2 were 6.2 (5.1-7.6) for mRNA-1388 25 µg, 53.8 (26.8-108.1) for mRNA-1388 50 µg, 92.8 (43.6-197.6) for mRNA-1388 100 µg, and 5.0 (not estimable) for placebo. Persistent humoral responses were observed up to 1 year after vaccination and remained higher than placebo in the 2 higher mRNA-1388 dose groups. The development of CHIKV-binding antibodies followed a similar trend as that observed with neutralizing antibodies. CONCLUSIONS: mRNA-1388, the first mRNA vaccine against CHIKV, was well tolerated and elicited substantial and long-lasting neutralizing antibody responses in healthy adult participants in a nonendemic region. CLINICALTRIALS: gov: NCT03325075.


Assuntos
Febre de Chikungunya , Vírus Chikungunya , Humanos , Adulto , Febre de Chikungunya/prevenção & controle , Vacinas Sintéticas , Anticorpos Neutralizantes , Anticorpos Antivirais , Imunogenicidade da Vacina , Método Duplo-Cego
2.
Front Immunol ; 14: 1161149, 2023.
Artigo em Inglês | MEDLINE | ID: covidwho-20237016

RESUMO

Mosquito-borne viral diseases are a group of viral illnesses that are predominantly transmitted by mosquitoes, including viruses from the Togaviridae and Flaviviridae families. In recent years, outbreaks caused by Dengue and Zika viruses from the Flaviviridae family, and Chikungunya virus from the Togaviridae family, have raised significant concerns for public health. However, there are currently no safe and effective vaccines available for these viruses, except for CYD-TDV, which has been licensed for Dengue virus. Efforts to control the transmission of COVID-19, such as home quarantine and travel restrictions, have somewhat limited the spread of mosquito-borne viral diseases. Several vaccine platforms, including inactivated vaccines, viral-vector vaccines, live attenuated vaccines, protein vaccines, and nucleic acid vaccines, are being developed to combat these viruses. This review analyzes the various vaccine platforms against Dengue, Zika, and Chikungunya viruses and provides valuable insights for responding to potential outbreaks.


Assuntos
COVID-19 , Vírus Chikungunya , Culicidae , Dengue , Vacinas Virais , Infecção por Zika virus , Zika virus , Animais , Humanos , Mosquitos Vetores , Infecção por Zika virus/epidemiologia , Infecção por Zika virus/prevenção & controle , Vacinas Atenuadas , Dengue/epidemiologia , Dengue/prevenção & controle , Desenvolvimento de Vacinas
3.
Viruses ; 15(5)2023 05 14.
Artigo em Inglês | MEDLINE | ID: covidwho-20232730

RESUMO

Chikungunya virus (CHIKV) and Zika virus (ZIKV) are important disease-causing agents worldwide. Currently, there are no antiviral drugs or vaccines approved to treat these viruses. However, peptides have shown great potential for new drug development. A recent study described (p-BthTX-I)2K [(KKYRYHLKPF)2K], a peptide derived from the Bothropstoxin-I toxin in the venom of the Bothrops jararacussu snake, showed antiviral activity against SARS-CoV-2. In this study, we assessed the activity of this peptide against CHIKV and ZIKV and its antiviral action in the different stages of the viral replication cycle in vitro. We observed that (p-BthTX-I)2K impaired CHIKV infection by interfering with the early steps of the viral replication cycle, reducing CHIKV entry into BHK-21 cells specifically by reducing both the attachment and internalization steps. (p-BthTX-I)2K also inhibited the ZIKV replicative cycle in Vero cells. The peptide protected the cells against ZIKV infection and decreased the levels of the viral RNA and the NS3 protein of this virus at viral post-entry steps. In conclusion, this study highlights the potential of the (p-BthTX-I)2K peptide to be a novel broad-spectrum antiviral candidate that targets different steps of the replication cycle of both CHIKV and ZIKV.


Assuntos
COVID-19 , Febre de Chikungunya , Vírus Chikungunya , Vírus , Infecção por Zika virus , Zika virus , Animais , Chlorocebus aethiops , Humanos , Infecção por Zika virus/tratamento farmacológico , Zika virus/genética , Células Vero , Antivirais/farmacologia , Antivirais/uso terapêutico , Replicação Viral , SARS-CoV-2 , Vírus Chikungunya/genética , Peptídeos/farmacologia , Peptídeos/uso terapêutico
4.
Front Immunol ; 14: 1129118, 2023.
Artigo em Inglês | MEDLINE | ID: covidwho-2298964

RESUMO

Chikungunya fever (CHIKF) has spread to more than 100 countries worldwide, with frequent outbreaks in Europe and the Americas in recent years. Despite the relatively low lethality of infection, patients can suffer from long-term sequelae. Until now, no available vaccines have been approved for use; however, increasing attention is being paid to the development of vaccines against chikungunya virus (CHIKV), and the World Health Organization has included vaccine development in the initial blueprint deliverables. Here, we developed an mRNA vaccine using the nucleotide sequence encoding structural proteins of CHIKV. And immunogenicity was evaluated by neutralization assay, Enzyme-linked immunospot assay and Intracellular cytokine staining. The results showed that the encoded proteins elicited high levels of neutralizing antibody titers and T cell-mediated cellular immune responses in mice. Moreover, compared with the wild-type vaccine, the codon-optimized vaccine elicited robust CD8+ T-cell responses and mild neutralizing antibody titers. In addition, higher levels of neutralizing antibody titers and T-cell immune responses were obtained using a homologous booster mRNA vaccine regimen of three different homologous or heterologous booster immunization strategies. Thus, this study provides assessment data to develop vaccine candidates and explore the effectiveness of the prime-boost approach.


Assuntos
Febre de Chikungunya , Vírus Chikungunya , Vacinas Virais , Animais , Camundongos , Vírus Chikungunya/genética , Vacinas Virais/genética , Anticorpos Antivirais , Anticorpos Neutralizantes
5.
Cell Rep Med ; 4(5): 101024, 2023 05 16.
Artigo em Inglês | MEDLINE | ID: covidwho-2295352

RESUMO

RNA viruses continue to remain a threat for potential pandemics due to their rapid evolution. Potentiating host antiviral pathways to prevent or limit viral infections is a promising strategy. Thus, by testing a library of innate immune agonists targeting pathogen recognition receptors, we observe that Toll-like receptor 3 (TLR3), stimulator of interferon genes (STING), TLR8, and Dectin-1 ligands inhibit arboviruses, Chikungunya virus (CHIKV), West Nile virus, and Zika virus to varying degrees. STING agonists (cAIMP, diABZI, and 2',3'-cGAMP) and Dectin-1 agonist scleroglucan demonstrate the most potent, broad-spectrum antiviral function. Furthermore, STING agonists inhibit severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and enterovirus-D68 (EV-D68) infection in cardiomyocytes. Transcriptome analysis reveals that cAIMP treatment rescue cells from CHIKV-induced dysregulation of cell repair, immune, and metabolic pathways. In addition, cAIMP provides protection against CHIKV in a chronic CHIKV-arthritis mouse model. Our study describes innate immune signaling circuits crucial for RNA virus replication and identifies broad-spectrum antivirals effective against multiple families of pandemic potential RNA viruses.


Assuntos
COVID-19 , Vírus Chikungunya , Vírus de RNA , Infecção por Zika virus , Zika virus , Animais , Camundongos , SARS-CoV-2 , Antivirais/farmacologia , Antivirais/uso terapêutico , Vírus Chikungunya/fisiologia , Imunidade Inata
6.
Lett Appl Microbiol ; 74(6): 992-1000, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: covidwho-2267626

RESUMO

Chikungunya is a fast-mutating virus causing Chikungunya virus disease (ChikvD) with a significant load of disability-adjusted life years (DALY) around the world. The outbreak of this virus is significantly higher in the tropical countries. Several experiments have identified crucial viral-host protein-protein interactions (PPIs) between Chikungunya Virus (Chikv) and the human host. However, no standard database that catalogs this PPI information exists. Here we develop a Chikv-Human PPI database, ChikvInt, to facilitate understanding ChikvD disease pathogenesis and the progress of vaccine studies. ChikvInt consists of 109 interactions and is available at www.chikvint.com.


Assuntos
Febre de Chikungunya , Vírus Chikungunya , Febre de Chikungunya/patologia , Humanos
7.
Viruses ; 14(9)2022 09 10.
Artigo em Inglês | MEDLINE | ID: covidwho-2143625

RESUMO

Chikungunya virus (CHIKV) is an arthropod-borne virus (arbovirus) transmitted by Aedes mosquitoes. The human infection usually manifests as a febrile and incapacitating arthritogenic illness, self-limiting and non-lethal. However, since 2013, CHIKV spreading through the tropics and to the Americas was accompanied by an increasing number of cases of atypical disease presentation, namely severe neuropathies and neonatal infection due to intrapartum vertical transmission. The pathophysiological mechanisms underlying these conditions have not been fully elucidated. However, arbovirus intrahost genetic diversity is thought to be linked to viral pathogenesis. To determine whether particular viral variants could be somehow associated, we analyzed the intrahost genetic diversity of CHIKV in three infected patients with neurological manifestations and three mothers infected during the intrapartum period, as well as their babies following vertical transmission. No statistically supported differences were observed for the genetic variability (nucleotide substitutions/gene length) along the genome between the groups. However, the newborn and cerebrospinal fluid samples (corresponding to virus passed through the placenta and/or the blood-brain barrier (BBB)) presented a different composition of their intrahost mutant ensembles compared to maternal or patient serum samples, even when concurrent. This finding could be consistent with the unidirectional virus transmission through these barriers, and the effect of selective bottlenecks during the transmission event. In addition, a higher proportion of defective variants (insertions/deletions and stop codons) was detected in the CSF and maternal samples and those were mainly distributed within the viral non-structural genes. Since defective viral genomes in RNA viruses are known to contribute to the outcome of acute viral infections and influence disease severity, their role in these atypical cases should be further investigated. Finally, with the in silico approach adopted, we detected no relevant non-conservative mutational pattern that could provide any hint of the pathophysiological mechanisms underlying these atypical cases. The present analysis represents a unique contribution to our understanding of the transmission events in these cases and generates hypotheses regarding underlying mechanisms, that can be explored further.


Assuntos
Aedes , Febre de Chikungunya , Vírus Chikungunya , Doenças Transmissíveis , Animais , Brasil/epidemiologia , Vírus Chikungunya/genética , Códon de Terminação , Humanos , Recém-Nascido , Nucleotídeos
8.
Molecules ; 27(16)2022 Aug 10.
Artigo em Inglês | MEDLINE | ID: covidwho-2023932

RESUMO

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.


Assuntos
Alcaloides , Infecções por Alphavirus , Vírus Chikungunya , Quinolizidinas , Alcaloides/farmacologia , Alcaloides/uso terapêutico , Infecções por Alphavirus/tratamento farmacológico , Infecções por Alphavirus/patologia , Antivirais/farmacologia , Antivirais/uso terapêutico , Vírus Chikungunya/fisiologia , Humanos , Indóis/uso terapêutico , Inflamação , Isoquinolinas , Quinolizidinas/farmacologia
10.
Vaccine ; 40(35): 5263-5274, 2022 08 19.
Artigo em Inglês | MEDLINE | ID: covidwho-1886119

RESUMO

Inactivated viral vaccines have long been used in humans for diseases of global health threat (e.g., poliomyelitis and pandemic and seasonal influenza) and the technology of inactivation has more recently been used for emerging diseases such as West Nile, Chikungunya, Ross River, SARS and especially for COVID-19. The Brighton Collaboration Benefit-Risk Assessment of VAccines by TechnolOgy (BRAVATO) Working Group has prepared standardized templates to describe the key considerations for the benefit and risk of several vaccine platform technologies, including inactivated viral vaccines. This paper uses the BRAVATO inactivated virus vaccine template to review the features of an inactivated whole chikungunya virus (CHIKV) vaccine that has been evaluated in several preclinical studies and clinical trials. The inactivated whole CHIKV vaccine was cultured on Vero cells and inactivated by ß-propiolactone. This provides an effective, flexible system for high-yield manufacturing. The inactivated whole CHIKV vaccine has favorable thermostability profiles, compatible with vaccine supply chains. Safety data are compiled in the current inactivated whole CHIKV vaccine safety database with unblinded data from the ongoing studies: 850 participants from phase II study (parts A and B) outside of India, and 600 participants from ongoing phase II study in India, and completed phase I clinical studies for 60 subjects. Overall, the inactivated whole CHIKV vaccine has been well tolerated, with no significant safety issues identified. Evaluation of the inactivated whole CHIKV vaccine is continuing, with 1410 participants vaccinated as of 20 April 2022. Extensive evaluation of immunogenicity in humans shows strong, durable humoral immune responses.


Assuntos
COVID-19 , Febre de Chikungunya , Vírus Chikungunya , Vacinas Virais , Animais , Anticorpos Antivirais , COVID-19/prevenção & controle , Febre de Chikungunya/prevenção & controle , Chlorocebus aethiops , Humanos , Medição de Risco , Vacinas de Produtos Inativados , Células Vero
11.
Sensors (Basel) ; 22(12)2022 Jun 10.
Artigo em Inglês | MEDLINE | ID: covidwho-1884320

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic is a worldwide health anxiety. The rapid dispersion of the infection globally results in unparalleled economic, social, and health impacts. The pathogen that causes COVID-19 is known as a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A fast and low-cost diagnosis method for COVID-19 disease can play an important role in controlling its proliferation. Near-infrared spectroscopy (NIRS) is a quick, non-destructive, non-invasive, and inexpensive technique for profiling the chemical and physical structures of a wide range of samples. Furthermore, the NIRS has the advantage of incorporating the internet of things (IoT) application for the effective control and treatment of the disease. In recent years, a significant advancement in instrumentation and spectral analysis methods has resulted in a remarkable impact on the NIRS applications, especially in the medical discipline. To date, NIRS has been applied as a technique for detecting various viruses including zika (ZIKV), chikungunya (CHIKV), influenza, hepatitis C, dengue (DENV), and human immunodeficiency (HIV). This review aims to outline some historical and contemporary applications of NIRS in virology and its merit as a novel diagnostic technique for SARS-CoV-2.


Assuntos
COVID-19 , Vírus Chikungunya , Vírus da Dengue , Dengue , Infecção por Zika virus , Zika virus , COVID-19/diagnóstico , Dengue/diagnóstico , Humanos , SARS-CoV-2 , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Infecção por Zika virus/diagnóstico
12.
mBio ; 13(3): e0073122, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: covidwho-1865140

RESUMO

Chikungunya virus (CHIKV) is an arthritogenic reemerging virus replicating in plasma membrane-derived compartments termed "spherules." Here, we identify the human transmembrane protein CD81 as host factor required for CHIKV replication. Ablation of CD81 results in decreased CHIKV permissiveness, while overexpression enhances infection. CD81 is dispensable for virus uptake but critically required for viral genome replication. Likewise, murine CD81 is crucial for CHIKV permissiveness and is expressed in target cells such as dermal fibroblasts, muscle and liver cells. Whereas related alphaviruses, including Ross River virus (RRV), Semliki Forest virus (SFV), Sindbis virus (SINV) and Venezuelan equine encephalitis virus (VEEV), also depend on CD81 for infection, RNA viruses from other families, such as coronaviruses, replicate independently of CD81. Strikingly, the replication-enhancing function of CD81 is linked to cholesterol binding. These results define a mechanism exploited by alphaviruses to hijack the membrane microdomain-modeling protein CD81 for virus replication through interaction with cholesterol. IMPORTANCE In this study, we discover the tetraspanin CD81 as a host factor for the globally emerging chikungunya virus and related alphaviruses. We show that CD81 promotes replication of viral genomes in human and mouse cells, while virus entry into cells is independent of CD81. This provides novel insights into how alphaviruses hijack host proteins to complete their life cycle. Alphaviruses replicate at distinct sites of the plasma membrane, which are enriched in cholesterol. We found that the cholesterol-binding ability of CD81 is important for its function as an alphavirus host factor. This discovery thus broadens our understanding of the alphavirus replication process and the use of host factors to reprogram cells into virus replication factories.


Assuntos
Febre de Chikungunya , Vírus Chikungunya , Vírus , Animais , Vírus Chikungunya/genética , Colesterol/metabolismo , Humanos , Camundongos , Tetraspaninas/metabolismo , Replicação Viral/genética , Vírus/metabolismo
13.
PLoS One ; 17(4): e0265820, 2022.
Artigo em Inglês | MEDLINE | ID: covidwho-1855004

RESUMO

INTRODUCTION: The rapid expansion of the novel SARS-CoV-2 virus has raised serious public health concerns due to the possibility of misdiagnosis in regions where arboviral diseases are endemic. We performed the first study in northern Peru to describe the detection of SARS-CoV-2 IgM antibodies in febrile patients with a suspected diagnosis of dengue and chikungunya fever. MATERIALS AND METHODS: A consecutive cross-sectional study was performed in febrile patients attending primary healthcare centers from April 2020 through March 2021. Patients enrolled underwent serum sample collection for the molecular and serological detection of DENV and CHIKV. Also, serological detection of IgM antibodies against SARS-CoV-2 was performed. RESULTS: 464 patients were included during the study period, of which (40.51%) were positive for one pathogen, meanwhile (6.90%) presented co-infections between 2 or more pathogens. The majority of patients with monoinfections were positive for SARS-CoV-2 IgM with (73.40%), followed by DENV 18.09% and CHIKV (8.51%). The most frequent co-infection was DENV + SARS-CoV-2 with (65.63%), followed by DENV + CHIKV and DENV + CHIKV + SARS-CoV-2, both with (12.50%). The presence of polyarthralgias in hands (43.75%, p<0.01) and feet (31.25%, p = 0.05) were more frequently reported in patients with CHIKV monoinfection. Also, conjunctivitis was more common in patients positive for SARS-CoV-2 IgM (11.45%, p<0.01). The rest of the symptoms were similar among all the study groups. CONCLUSION: SARS-CoV-2 IgM antibodies were frequently detected in acute sera from febrile patients with a clinical suspicion of arboviral disease. The presence of polyarthralgias in hands and feet may be suggestive of CHIKV infection. These results reaffirm the need to consider SARS-CoV-2 infection as a main differential diagnosis of acute febrile illness in arboviruses endemic areas, as well as to consider co-infections between these pathogens.


Assuntos
COVID-19 , Febre de Chikungunya , Vírus Chikungunya , Coinfecção , Vírus da Dengue , Dengue , Infecção por Zika virus , Anticorpos Antivirais , Artralgia , COVID-19/diagnóstico , COVID-19/epidemiologia , Febre de Chikungunya/diagnóstico , Febre de Chikungunya/epidemiologia , Coinfecção/diagnóstico , Coinfecção/epidemiologia , Estudos Transversais , Dengue/diagnóstico , Dengue/epidemiologia , Febre/diagnóstico , Humanos , Imunoglobulina M , Peru/epidemiologia , SARS-CoV-2 , Infecção por Zika virus/epidemiologia
14.
Infect Genet Evol ; 99: 105243, 2022 04.
Artigo em Inglês | MEDLINE | ID: covidwho-1763893

RESUMO

Mayaro Virus is an emerging arbovirus which can be responsible of important outbreaks in tropical regions. A retrospective study was performed in French Guiana, an ultraperipheral region of Europe in Amazonia. We identified 17 human cases between 2003 and 2019. The clinical and biological picture was close to Chikungunya with fever and arthralgia. One patient had acute meningo-encephalitis, and 4 had persistent arthralgia. Physicians should be aware of this virus, as imported cases in Europe have already occurred. AUTHOR SUMMARY: Latin America has experienced several epidemics of arboviruses in recent years, some known for a long time, such as the dengue virus, and others of more recent introduction such as the chikungunya or Zika viruses. There are other arboviruses for the moment more discreet which are rife with low noise in several countries of the continent, such as the Mayaro virus. This alphavirus, with a presentation similar to that of the chikungunya virus, is currently confined to transmission by forest mosquitoes, but its potential to be transmitted by coastal mosquitoes such as Aedes aegypti, make it a potential candidate for a continent-wide epidemic. It therefore seems necessary to know this virus as well as possible in order to anticipate the occurrence of a possible new epidemic. We present here a both demographic and clinical study of this endemic arbovirus disease in French Guiana.


Assuntos
Aedes , Arbovírus , Febre de Chikungunya , Vírus Chikungunya , Infecção por Zika virus , Zika virus , Animais , Artralgia , Estudos Transversais , Guiana Francesa/epidemiologia , Humanos , Mosquitos Vetores , Estudos Retrospectivos , Infecção por Zika virus/epidemiologia
15.
Clin Med (Lond) ; 22(1): 18-20, 2022 01.
Artigo em Inglês | MEDLINE | ID: covidwho-1737354

RESUMO

A large majority of neurological infections remain undiagnosed worldwide. Emerging and re-emerging infections are likely to be responsible for a significant proportion of these. Over the last two decades, several new organisms producing neurological infection and the neurotropic potential of many other known pathogens have been identified. Large outbreaks caused by re-emerging pathogens such as Chikungunya virus, Zika virus and Ebola virus have led to better delineation of their neurological manifestations. Recognition of the pandemic potential of emerging pathogens and an improved understanding of their host-vector-environment interactions would help us be better prepared to meet these emerging threats.


Assuntos
Febre de Chikungunya , Vírus Chikungunya , Doenças Transmissíveis Emergentes , Infecção por Zika virus , Zika virus , Febre de Chikungunya/diagnóstico , Febre de Chikungunya/epidemiologia , Doenças Transmissíveis Emergentes/diagnóstico , Doenças Transmissíveis Emergentes/epidemiologia , Surtos de Doenças , Humanos , Infecção por Zika virus/complicações , Infecção por Zika virus/diagnóstico , Infecção por Zika virus/epidemiologia
16.
Vector Borne Zoonotic Dis ; 21(11): 900-909, 2021 11.
Artigo em Inglês | MEDLINE | ID: covidwho-1532426

RESUMO

Background: A wide range of insect-specific viruses (ISVs) have been reported worldwide. There are no studies from India that have reported ISVs. The current study describes the identification of Phasi Charoen-like virus (PCLV) from Aedes aegypti mosquito-pools from six districts of Karnataka state, India. Materials and Methods: During the Chikungunya virus (CHIKV) outbreak in the Bangalore Urban district in 2019, using conventional PCR, it was found that both human and mosquito samples were positive for CHIKV. For retrieve the complete genome sequence, mosquito samples were subjected to next generation sequencing (NGS) analysis and PCLV was also found. During 2019, as part of a vector-borne disease surveillance, we received 50 mosquito pool samples from 6 districts of the state, all of them were subjected to NGS to identify PCLV. Results: The A. aegypti mosquito-pools samples were subjected to the NGS platform that led to identification of an ISV, PCLV. PCLV was identified in 26 A. aegypti mosquito-pools collected from 6 districts. We also found mixed infection of PCLV with the Dengue virus (DENV; genotypes 1 and 3) and CHIKV from five pools. The nucleotide identity for the L gene of Indian PCLV sequences ranged between 97.1% and 98.3% in comparison with the Thailand sequences. Conclusions: To the best of our knowledge, this is the first report of PCLV dual infection with DENV and CHIKV in India. The present study confirms the presence of PCLV in A. aegypti mosquitoes from Karnataka state. The study adds India in the global geographical distribution of PCLV.


Assuntos
Aedes , Vírus Chikungunya , Vírus de RNA , Animais , Vírus Chikungunya/genética , Índia/epidemiologia , Mosquitos Vetores
17.
Viruses ; 13(8)2021 08 12.
Artigo em Inglês | MEDLINE | ID: covidwho-1436097

RESUMO

Type III interferons (lambda IFNs) are a quite new, small family of three closely related cytokines with interferon-like activity. Attention to IFN-λ is mainly focused on direct antiviral activity in which, as with IFN-α, viral genome replication is inhibited without the participation of immune system cells. The heterodimeric receptor for lambda interferons is exposed mainly on epithelial cells, which limits its possible action on other cells, thus reducing the likelihood of developing undesirable side effects compared to type I IFN. In this study, we examined the antiviral potential of exogenous human IFN-λ1 in cellular models of viral infection. To study the protective effects of IFN-λ1, three administration schemes were used: 'preventive' (pretreatment); 'preventive/therapeutic' (pre/post); and 'therapeutic' (post). Three IFN-λ1 concentrations (from 10 to 500 ng/mL) were used. We have shown that human IFN-λ1 restricts SARS-CoV-2 replication in Vero cells with all three treatment schemes. In addition, we have shown a decrease in the viral loads of CHIKV and IVA with the 'preventive' and 'preventive/therapeutic' regimes. No significant antiviral effect of IFN-λ1 against AdV was detected. Our study highlights the potential for using IFN-λ as a broad-spectrum therapeutic agent against respiratory RNA viruses.


Assuntos
Adenovírus Humanos/efeitos dos fármacos , Vírus Chikungunya/efeitos dos fármacos , Vírus da Influenza A/efeitos dos fármacos , Interferons/farmacologia , SARS-CoV-2/efeitos dos fármacos , Células A549 , Adenovírus Humanos/fisiologia , Animais , Vírus Chikungunya/fisiologia , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Humanos , Vírus da Influenza A/fisiologia , Interferons/uso terapêutico , Interleucinas , Infecções por Vírus de RNA/tratamento farmacológico , Infecções por Vírus de RNA/prevenção & controle , Proteínas Recombinantes/farmacologia , SARS-CoV-2/fisiologia , Células Vero , Carga Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Interferon lambda
18.
Biomolecules ; 11(5)2021 05 18.
Artigo em Inglês | MEDLINE | ID: covidwho-1389275

RESUMO

Several RNA viruses, including SARS-CoV-2, can infect or use the eye as an entry portal to cause ocular or systemic diseases. Povidone-Iodine (PVP-I) is routinely used during ocular surgeries and eye banking as a cost-effective disinfectant due to its broad-spectrum antimicrobial activity, including against viruses. However, whether PVP-I can exert antiviral activities in virus-infected cells remains elusive. In this study, using Zika (ZIKV) and Chikungunya (CHIKV) virus infection of human corneal and retinal pigment epithelial cells, we report antiviral mechanisms of PVP-I. Our data showed that PVP-I, even at the lowest concentration (0.01%), drastically reduced viral replication in corneal and retinal cells without causing cellular toxicity. Antiviral effects of PVP-I against ZIKV and CHIKV were mediated by direct viral inactivation, thus attenuating the ability of the virus to infect host cells. Moreover, one-minute PVP-I exposure of infected ocular cells drastically reduced viral replication and the production of infectious progeny virions. Furthermore, viral-induced (CHIKV) expression of inflammatory genes (TNF-α, IL-6, IL-8, and IL1ß) were markedly reduced in PVP-I treated corneal epithelial cells. Together, our results demonstrate potent antiviral effects of PVP-I against ZIKV and CHIKV infection of ocular cells. Thus, a low dose of PVP-I can be used during tissue harvesting for corneal transplants to prevent potential transmission of RNA viruses via infected cells.


Assuntos
Antivirais/farmacologia , Povidona-Iodo/farmacologia , Vírus de RNA/fisiologia , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Vírus Chikungunya/fisiologia , Chlorocebus aethiops , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/virologia , SARS-CoV-2/fisiologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Células Vero , Zika virus/fisiologia
19.
Nat Commun ; 12(1): 4636, 2021 07 30.
Artigo em Inglês | MEDLINE | ID: covidwho-1347938

RESUMO

Chikungunya virus (CHIKV) is a reemerging mosquito-borne virus that causes swift outbreaks. Major concerns are the persistent and disabling polyarthralgia in infected individuals. Here we present the results from a first-in-human trial of the candidate simian adenovirus vectored vaccine ChAdOx1 Chik, expressing the CHIKV full-length structural polyprotein (Capsid, E3, E2, 6k and E1). 24 adult healthy volunteers aged 18-50 years, were recruited in a dose escalation, open-label, nonrandomized and uncontrolled phase 1 trial (registry NCT03590392). Participants received a single intramuscular injection of ChAdOx1 Chik at one of the three preestablished dosages and were followed-up for 6 months. The primary objective was to assess safety and tolerability of ChAdOx1 Chik. The secondary objective was to assess the humoral and cellular immunogenicity. ChAdOx1 Chik was safe at all doses tested with no serious adverse reactions reported. The vast majority of solicited adverse events were mild or moderate, and self-limiting in nature. A single dose induced IgG and T-cell responses against the CHIKV structural antigens. Broadly neutralizing antibodies against the four CHIKV lineages were found in all participants and as early as 2 weeks after vaccination. In summary, ChAdOx1 Chik showed excellent safety, tolerability and 100% PRNT50 seroconversion after a single dose.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Febre de Chikungunya/imunologia , Vírus Chikungunya/imunologia , Vacinas Virais/imunologia , Adolescente , Adulto , Febre de Chikungunya/prevenção & controle , Febre de Chikungunya/virologia , Vírus Chikungunya/classificação , Vírus Chikungunya/fisiologia , Citocinas/imunologia , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Fadiga/induzido quimicamente , Feminino , Cefaleia/induzido quimicamente , Humanos , Imunoglobulina G/imunologia , Injeções Intramusculares , Masculino , Pessoa de Meia-Idade , Linfócitos T/imunologia , Linfócitos T/metabolismo , Vacinação/métodos , Vacinas Virais/administração & dosagem , Vacinas Virais/efeitos adversos , Adulto Jovem
20.
Cells ; 10(5)2021 05 11.
Artigo em Inglês | MEDLINE | ID: covidwho-1274611

RESUMO

Th17 cells are recognized as indispensable in inducing protective immunity against bacteria and fungi, as they promote the integrity of mucosal epithelial barriers. It is believed that Th17 cells also play a central role in the induction of autoimmune diseases. Recent advances have evaluated Th17 effector functions during viral infections, including their critical role in the production and induction of pro-inflammatory cytokines and in the recruitment and activation of other immune cells. Thus, Th17 is involved in the induction both of pathogenicity and immunoprotective mechanisms seen in the host's immune response against viruses. However, certain Th17 cells can also modulate immune responses, since they can secrete immunosuppressive factors, such as IL-10; these cells are called non-pathogenic Th17 cells. Here, we present a brief review of Th17 cells and highlight their involvement in some virus infections. We cover these notions by highlighting the role of Th17 cells in regulating the protective and pathogenic immune response in the context of viral infections. In addition, we will be describing myocarditis and multiple sclerosis as examples of immune diseases triggered by viral infections, in which we will discuss further the roles of Th17 cells in the induction of tissue damage.


Assuntos
Miocardite/imunologia , Células Th17/metabolismo , Viroses/imunologia , Adenoviridae , Animais , Doenças Autoimunes/imunologia , Vírus Chikungunya , Citocinas/imunologia , Vírus da Dengue , Humanos , Sistema Imunitário , Imunossupressores/farmacologia , Inflamação , Interleucina-10/biossíntese , Linfócitos/citologia , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/virologia , Miocardite/metabolismo , Miocardite/virologia , Orthomyxoviridae , SARS-CoV-2 , Simplexvirus , Células Th1/citologia , Células Th2/citologia , Viroses/tratamento farmacológico , Viroses/metabolismo , Zika virus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA