Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
J Med Virol ; 96(7): e29787, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38988177

RESUMO

Chronic hepatitis C virus infection (HCV) causes liver inflammation and fibrosis, leading to the development of severe liver disease, such as cirrhosis or hepatocellular carcinoma (HCC). Approval of direct-acting antiviral drug combinations has revolutionized chronic HCV therapy, with virus eradication in >98% of the treated patients. The efficacy of these treatments is such that it is formally possible for cured patients to carry formerly infected cells that display irreversible transcriptional alterations directly caused by chronic HCV Infection. Combining differential transcriptomes from two different persistent infection models, we observed a major reversion of infection-related transcripts after complete infection elimination. However, a small number of transcripts were abnormally expressed in formerly infected cells. Comparison of the results obtained in proliferating and growth-arrested cell culture models suggest that permanent transcriptional alterations may be established by several mechanisms. Interestingly, some of these alterations were also observed in the liver biopsies of virologically cured patients. Overall, our data suggest a direct and permanent impact of persistent HCV infection on the host cell transcriptome even after virus elimination, possibly contributing to the development of HCC.


Assuntos
Antivirais , Hepacivirus , Hepatite C Crônica , Humanos , Antivirais/farmacologia , Antivirais/uso terapêutico , Hepacivirus/genética , Hepacivirus/efeitos dos fármacos , Hepatite C Crônica/tratamento farmacológico , Hepatite C Crônica/virologia , Transcriptoma , Infecção Persistente/virologia , Perfilação da Expressão Gênica , Fígado/virologia , Fígado/patologia , Carcinoma Hepatocelular/virologia , Transcrição Gênica/efeitos dos fármacos
2.
Virol J ; 21(1): 133, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38844960

RESUMO

BACKGROUND: Early diagnosis and treatment of HPV persistent infection and cervical intraepithelial neoplasia, which have yet to be thoroughly characterized in Guangxi, Southwestern China, are the key preventative measures for the development of cervical cancer in women, particularly in HIV-infected women. METHODS: A retrospective study of 181 patients with HPV infection or cervical intraepithelial neoplasia who received surgical excision of lesions and were prospectively enrolled at the Fourth People's Hospital of Nanning between January 2018 and February 2023 was performed. HPV-infected patients were divided into two subgroups: HIV-infected and HIV/HPV-coinfected patients and compare differences between these groups. RESULTS: HPV16, 18, 52, and 58 were the most prevalent HPV genotypes. High-risk HPV was significantly co-infected with multiple genotypes (P = 0.0332). HIV-infected women were predisposed to HPV infection (P < 0.0001), and the development of cervical cancer at a young age (P = 0.0336) compared to HIV-uninfected women and the loop electrosurgical excision procedure (P = 0.0480) is preferred for the treatment. CONCLUSIONS: HIV infection may increase HPV prevalence and lead to cervical cancer development at a young age. The loop electrosurgical excision procedure is an efficient evaluation and treatment strategy for HIV-infected women suffering from cervical intraepithelial neoplasia.


Assuntos
Coinfecção , Infecções por HIV , Infecções por Papillomavirus , Displasia do Colo do Útero , Neoplasias do Colo do Útero , Humanos , Feminino , Infecções por HIV/complicações , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/virologia , Estudos Retrospectivos , Adulto , Pessoa de Meia-Idade , Displasia do Colo do Útero/virologia , Displasia do Colo do Útero/cirurgia , Displasia do Colo do Útero/complicações , Neoplasias do Colo do Útero/virologia , Neoplasias do Colo do Útero/cirurgia , Coinfecção/virologia , China/epidemiologia , Genótipo , Prevalência , Papillomaviridae/genética , Papillomaviridae/isolamento & purificação , Infecção Persistente/virologia , Adulto Jovem
3.
J Med Virol ; 96(6): e29753, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38895800

RESUMO

Human papillomavirus (HPV) type 81 has recently become one of the most common low-risk HPV types; however, literature focusing on it is limited. This study aimed to analyze the reasons for the increased detection rate of HPV81 and investigate its evolving pathogenicity. We analyzed the detection rates and trends of HPV81 in 229 061 exfoliated cervical cell samples collected from 2014 to 2023; collected samples of HPV81 single infections from two different time periods; and analyzed the allele frequencies, positive selection, viral load, persistent infection capacity, and pathogenicity of E6 and E7 genotypes. We found that the detection rate of HPV81 ranked first among the low-risk types in exfoliated cervical cells and exhibited a significantly increasing trend (p < 0.001). The frequency of the E6 prototype allele of HPV81 (n = 317) was significantly increased (p = 0.018) and demonstrated the strongest adaptive capacity. The viral load and persistent infection capacity of the E6 prototype were significantly higher than those of the mutants, thus serving as key drivers for increasing the detection rate of HPV81 and enhancing its pathogenicity. The viral load was positively correlated with persistent infection capacity and pathogenicity. Persistent infection was a crucial factor in the pathogenicity of HPV81. Successful adaptive evolution of HPV81 is accompanied by enhanced pathogenicity.


Assuntos
Genótipo , Infecções por Papillomavirus , Infecção Persistente , Polimorfismo Genético , Carga Viral , Humanos , Infecções por Papillomavirus/virologia , Feminino , Infecção Persistente/virologia , Colo do Útero/virologia , Colo do Útero/patologia , Adulto , Papillomaviridae/genética , Papillomaviridae/patogenicidade , Papillomaviridae/classificação , Papillomaviridae/isolamento & purificação , Frequência do Gene , Proteínas Oncogênicas Virais/genética , Virulência/genética , Alphapapillomavirus/genética , Alphapapillomavirus/patogenicidade , Alphapapillomavirus/classificação , Alphapapillomavirus/isolamento & purificação , Papillomavirus Humano
4.
PLoS Pathog ; 20(5): e1011961, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38701091

RESUMO

Noroviruses (NoVs) are a leading cause of viral gastroenteritis. Despite global clinical relevance, our understanding of how host factors, such as antiviral cytokines interferons (IFNs), modulate NoV population dynamics is limited. Murine NoV (MNoV) is a tractable in vivo model for the study of host regulation of NoV. A persistent strain of MNoV, CR6, establishes a reservoir in intestinal tuft cells for chronic viral shedding in stool. However, the influence of host innate immunity and permissive cell numbers on viral population dynamics is an open question. We generated a pool of 20 different barcoded viruses (CR6BC) by inserting 6-nucleotide barcodes at the 3' position of the NS4 gene and used this pool as our viral inoculum for in vivo infections of different mouse lines. We found that over the course of persistent CR6 infection, shed virus was predominantly colon-derived, and viral barcode richness decreased over time irrespective of host immune status, suggesting that persistent infection involves a series of reinfection events. In mice lacking the IFN-λ receptor, intestinal barcode richness was enhanced, correlating with increased viral intestinal replication. IL-4 treatment, which increases tuft cell numbers, also increased barcode richness, indicating the abundance of permissive tuft cells to be a bottleneck during CR6 infection. In mice lacking type I IFN signaling (Ifnar1-/-) or all IFN signaling (Stat1-/-), barcode diversity at extraintestinal sites was dramatically increased, implicating different IFNs as critical bottlenecks at specific tissue sites. Of interest, extraintestinal barcodes were overlapping but distinct from intestinal barcodes, indicating that disseminated virus represents a distinct viral population than that replicating in the intestine. Barcoded viruses are a valuable tool to explore the influence of host factors on viral diversity in the context of establishment and maintenance of infection as well as dissemination and have provided important insights into how NoV infection proceeds in immunocompetent and immunocompromised hosts.


Assuntos
Infecções por Caliciviridae , Interferons , Norovirus , Animais , Norovirus/fisiologia , Infecções por Caliciviridae/virologia , Infecções por Caliciviridae/imunologia , Camundongos , Interferons/metabolismo , Infecção Persistente/virologia , Infecção Persistente/imunologia , Camundongos Endogâmicos C57BL , Mucosa Intestinal/virologia , Mucosa Intestinal/imunologia , Gastroenterite/virologia , Replicação Viral , Camundongos Knockout , Imunidade Inata , Eliminação de Partículas Virais
5.
Oncologist ; 29(6): 457-464, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38630576

RESUMO

Human papillomavirus (HPV)-associated malignancies account for ~5% of human cancers worldwide. Thirteen, or more, HPV types are oncogenic, but infection with these viruses is common and usually cleared within 2 years. Only infections that become persistent are associated with the development of cancer, often occurring several decades later. These cancers mostly arise in 6 different anatomical regions: 5 are anogenital (anus, cervix, penis, vagina, and vulva) and the sixth is the oropharynx. Oncogenic HPVs promote cellular proliferation and genomic instability, but the anatomical niche of the target tissue also plays an important role in the development of cancer. Cells that reside in transitional regions between different types of epithelia, such as in the anus, cervix, and oropharynx, are particularly vulnerable to oncogenesis.


Assuntos
Infecções por Papillomavirus , Humanos , Infecções por Papillomavirus/virologia , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/patologia , Feminino , Masculino , Papillomaviridae/patogenicidade , Neoplasias/virologia , Neoplasias/patologia , Neoplasias/complicações , Infecção Persistente/virologia
6.
Emerg Microbes Infect ; 13(1): 2348526, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38683015

RESUMO

The foot-and-mouth disease virus (FMDV) Leader proteinase Lpro inhibits host mRNA translation and blocks the interferon response which promotes viral survival. Lpro is not required for viral replication in vitro but serotype A FMDV lacking Lpro has been shown to be attenuated in cattle and pigs. However, it is not known, whether leaderless viruses can cause persistent infection in vivo after simulated natural infection and whether the attenuated phenotype is the same in other serotypes. We have generated an FMDV O/FRA/1/2001 variant lacking most of the Lpro coding region (ΔLb). Cattle were inoculated intranasopharyngeally and observed for 35 days to determine if O FRA/1/2001 ΔLb is attenuated during the acute phase of infection and whether it can maintain a persistent infection in the upper respiratory tract. We found that although this leaderless virus can replicate in vitro in different cell lines, it is unable to establish an acute infection with vesicular lesions and viral shedding nor is it able to persistently infect bovine pharyngeal tissues.


Assuntos
Doenças dos Bovinos , Vírus da Febre Aftosa , Febre Aftosa , Infecção Persistente , Sorogrupo , Replicação Viral , Animais , Bovinos , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/fisiologia , Vírus da Febre Aftosa/classificação , Vírus da Febre Aftosa/patogenicidade , Vírus da Febre Aftosa/isolamento & purificação , Febre Aftosa/virologia , Doenças dos Bovinos/virologia , Infecção Persistente/virologia , Linhagem Celular , Endopeptidases/genética , Endopeptidases/metabolismo , Eliminação de Partículas Virais
7.
Microbiol Immunol ; 68(5): 179-184, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38433377

RESUMO

BK polyomavirus (BKPyV) was the first human polyomavirus to be isolated from an immunosuppressed kidney transplant recipient in 1971. BKPyV reactivation causes BKPyV-associated nephropathy and hemorrhagic cystitis. However, the mechanisms underlying BKPyV replication remain unclear. In the present study, we performed the long-term cultivation of COS-7 cells transfected with archetype KOM-5 DNA, which were designated as COS-BK cells. BKPyV derived from COS-BK cells was characterized by analyzing the amount of the virus based on hemagglutination, viral replication, and the production of viral protein 1 (VP1). Immunostaining showed that VP1-positive cells accounted for a small percentage of COS-BK cells. The nucleotide sequences encompassing the origin of the DNA replication of BKPyV derived from COS-BK cells were generated from KOM-5 by the deletion of an 8-bp sequence, which did not involve T antigen binding sites. BKPyV replicated most efficiently in COS-BK cells in DMEM containing 2% fetal bovine serum. These results indicate that COS-BK cells are a suitable culture system for studying the persistent infection of archetype BKPyV.


Assuntos
Vírus BK , Infecções por Polyomavirus , Replicação Viral , Vírus BK/fisiologia , Vírus BK/genética , Animais , Chlorocebus aethiops , Células COS , Infecções por Polyomavirus/virologia , Humanos , Proteínas do Capsídeo/genética , DNA Viral/genética , Infecção Persistente/virologia , Antígenos Virais de Tumores/genética , Infecções Tumorais por Vírus/virologia
8.
Nature ; 626(8001): 1094-1101, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38383783

RESUMO

Persistent SARS-CoV-2 infections may act as viral reservoirs that could seed future outbreaks1-5, give rise to highly divergent lineages6-8 and contribute to cases with post-acute COVID-19 sequelae (long COVID)9,10. However, the population prevalence of persistent infections, their viral load kinetics and evolutionary dynamics over the course of infections remain largely unknown. Here, using viral sequence data collected as part of a national infection survey, we identified 381 individuals with SARS-CoV-2 RNA at high titre persisting for at least 30 days, of which 54 had viral RNA persisting at least 60 days. We refer to these as 'persistent infections' as available evidence suggests that they represent ongoing viral replication, although the persistence of non-replicating RNA cannot be ruled out in all. Individuals with persistent infection had more than 50% higher odds of self-reporting long COVID than individuals with non-persistent infection. We estimate that 0.1-0.5% of infections may become persistent with typically rebounding high viral loads and last for at least 60 days. In some individuals, we identified many viral amino acid substitutions, indicating periods of strong positive selection, whereas others had no consensus change in the sequences for prolonged periods, consistent with weak selection. Substitutions included mutations that are lineage defining for SARS-CoV-2 variants, at target sites for monoclonal antibodies and/or are commonly found in immunocompromised people11-14. This work has profound implications for understanding and characterizing SARS-CoV-2 infection, epidemiology and evolution.


Assuntos
COVID-19 , Inquéritos Epidemiológicos , Infecção Persistente , SARS-CoV-2 , Humanos , Substituição de Aminoácidos , Anticorpos Monoclonais/imunologia , COVID-19/epidemiologia , COVID-19/virologia , Evolução Molecular , Hospedeiro Imunocomprometido/imunologia , Mutação , Infecção Persistente/epidemiologia , Infecção Persistente/virologia , Síndrome de COVID-19 Pós-Aguda/epidemiologia , Síndrome de COVID-19 Pós-Aguda/virologia , Prevalência , RNA Viral/análise , RNA Viral/genética , SARS-CoV-2/química , SARS-CoV-2/classificação , SARS-CoV-2/genética , SARS-CoV-2/imunologia , SARS-CoV-2/isolamento & purificação , Seleção Genética , Autorrelato , Fatores de Tempo , Carga Viral , Replicação Viral
9.
J Virol ; 97(12): e0100823, 2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-37962378

RESUMO

IMPORTANCE: The human gammaherpesviruses Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus are etiologic agents of numerous B cell lymphomas. A hallmark of gammaherpesvirus infection is their ability to establish lifelong latency in B cells. However, the specific mechanisms that mediate chronic infection in B cells in vivo remain elusive. Cellular E3 ubiquitin ligases regulate numerous biological processes by catalyzing ubiquitylation and modifying protein location, function, or half-life. Many viruses hijack host ubiquitin ligases to evade antiviral host defense and promote viral fitness. Here, we used the murine gammaherpesvirus 68 in vivo system to demonstrate that the E3 ligase Cul4b is essential for this virus to establish latency in germinal center B cells. These findings highlight an essential role for this E3 ligase in promoting chronic gammaherpesvirus infection in vivo and suggest that targeted inhibition of E3 ligases may provide a novel and effective intervention strategy against gammaherpesvirus-associated diseases.


Assuntos
Linfócitos B , Gammaherpesvirinae , Infecções por Herpesviridae , Infecção Persistente , Animais , Camundongos , Linfócitos B/enzimologia , Linfócitos B/metabolismo , Linfócitos B/virologia , Proteínas Culina/metabolismo , Gammaherpesvirinae/fisiologia , Centro Germinativo/citologia , Centro Germinativo/virologia , Infecções por Herpesviridae/enzimologia , Infecções por Herpesviridae/virologia , Infecção Persistente/enzimologia , Infecção Persistente/virologia , Ubiquitinas/metabolismo , Latência Viral
10.
Proc Natl Acad Sci U S A ; 119(35): e2205037119, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35994644

RESUMO

Viruses are important ecological, biogeochemical, and evolutionary drivers in every environment. Upon infection, they often cause the lysis of the host cell. However, some viruses exhibit alternative life cycles, such as chronic infections without cell lysis. The nature and the impact of chronic infections in prokaryotic host organisms remains largely unknown. Here, we characterize a novel haloarchaeal virus, Haloferax volcanii pleomorphic virus 1 (HFPV-1), which is currently the only virus infecting the model haloarchaeon Haloferax volcanii DS2, and demonstrate that HFPV-1 and H. volcanii are a great model system to study virus-host interactions in archaea. HFPV-1 is a pleomorphic virus that causes a chronic infection with continuous release of virus particles, but host and virus coexist without cell lysis or the appearance of resistant cells. Despite an only minor impact of the infection on host growth, we uncovered an extensive remodeling of the transcriptional program of the host (up to 1,049 differentially expressed genes). These changes are highlighted by a down-regulation of two endogenous provirus regions in the host genome, and we show that HFPV-1 infection is strongly influenced by a cross-talk between HFPV-1 and one of the proviruses mediated by a superinfection-like exclusion mechanism. Furthermore, HFPV-1 has a surprisingly wide host range among haloarchaea, and purified virus DNA can cause an infection after transformation into the host, making HFPV-1 a candidate for being developed into a genetic tool for a range of so far inaccessible haloarchaea.


Assuntos
Proteínas Arqueais , Haloferax volcanii , Interações entre Hospedeiro e Microrganismos , Infecção Persistente , Provírus , Viroses , Proteínas Arqueais/metabolismo , Genoma , Haloferax volcanii/genética , Haloferax volcanii/metabolismo , Haloferax volcanii/virologia , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Infecção Persistente/terapia , Infecção Persistente/virologia , Provírus/genética , Provírus/isolamento & purificação , Provírus/metabolismo , Viroses/metabolismo , Viroses/virologia
11.
J Virol ; 96(11): e0044222, 2022 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-35546119

RESUMO

Interferons (IFNs) are cytokines that induce a global change in the cell to establish antiviral immunity. We previously demonstrated that human adenovirus (HAdV) exploits IFN-induced viral repression to persist in infected cells. Although this in vitro persistence model has been described, the mechanism behind how persistent HAdV infection is established is not well understood. In this study, we demonstrate that IFN signaling is essential for viral repression and promoting persistent infection. Cyclin-dependent kinase 4 (CDK4), an antagonist of retinoblastoma (Rb) family proteins, was shown to disrupt the viral repression induced by IFNs. Consistent with this result, knockout of the Rb family proteins pRb, p107, and/or p130 drastically reduced the effect of IFNs on viral replication. The pRb protein specifically contributed the greatest effect to IFN inhibition of viral replication. Interestingly, IFNs did not impact pRb through direct changes in protein or phosphorylation levels. Cells treated with IFNs continued to cycle normally, consistent with observations that persistently infected cells remain for long periods of time in the host and in our in vitro persistent infection model. Finally, we observed that histone deacetylase (HDAC) inhibitors activated productive viral replication in persistently infected cells in the presence of IFN. Thus, HDACs, specifically class I HDACs, which are commonly associated with Rb family proteins, play a major role in the maintenance of persistent HAdV infection in vitro. This study uncovers the critical role of pRb and class I HDACs in the IFN-induced formation of a repressor complex that promotes persistent HAdV infections. IMPORTANCE Adenoviruses are ubiquitous viruses infecting more than 90% of the human population. HAdVs cause persistent infections that may lead to serious complications in immunocompromised patients. Therefore, exploring how HAdVs establish persistent infections is critical for understanding viral reactivation in immunosuppressed individuals. The mechanism underlying HAdV persistence has not been fully explored. Here, we provide insight into the contributions of the host cell to IFN-mediated persistent HAdV infection. We found that HAdV-C5 productive infection is inhibited by an Rb-E2F-HDAC repressor complex. Treatment with HDAC inhibitors converted a persistent infection to a lytic infection. Our results suggest that this process involves the noncanonical regulation of Rb-E2F signaling. This study provides insight into a highly prevalent human pathogen, bringing a new level of complexity and understanding to the replicative cycle.


Assuntos
Infecções por Adenovirus Humanos , Adenovírus Humanos , Interferons , Infecção Persistente , Infecções por Adenovirus Humanos/imunologia , Adenovírus Humanos/fisiologia , Fatores de Transcrição E2F/imunologia , Histona Desacetilases/imunologia , Humanos , Interferons/imunologia , Infecção Persistente/imunologia , Infecção Persistente/virologia , Proteína do Retinoblastoma/imunologia
12.
Viruses ; 14(2)2022 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-35215808

RESUMO

Human Papillomaviruses have co-evolved with their human host, with each of the over 200 known HPV types infecting distinct epithelial niches to cause diverse disease pathologies. Despite the success of prophylactic vaccines in preventing high-risk HPV infection, the development of HPV anti-viral therapies has been hampered by the lack of enzymatic viral functions, and by difficulties in translating the results of in vitro experiments into clinically useful treatment regimes. In this review, we discuss recent advances in anti-HPV drug development, and highlight the importance of understanding persistent HPV infections for future anti-viral design. In the infected epithelial basal layer, HPV genomes are maintained at a very low copy number, with only limited viral gene expression; factors which allow them to hide from the host immune system. However, HPV gene expression confers an elevated proliferative potential, a delayed commitment to differentiation, and preferential persistence of the infected cell in the epithelial basal layer, when compared to their uninfected neighbours. To a large extent, this is driven by the viral E6 protein, which functions in the HPV life cycle as a modulator of epithelial homeostasis. By targeting HPV gene products involved in the maintenance of the viral reservoir, there appears to be new opportunities for the control or elimination of chronic HPV infections.


Assuntos
Alphapapillomavirus/efeitos dos fármacos , Antivirais/uso terapêutico , Infecções por Papillomavirus/tratamento farmacológico , Infecção Persistente/tratamento farmacológico , Antivirais/farmacologia , Desenvolvimento de Medicamentos , Epitélio/efeitos dos fármacos , Epitélio/patologia , Epitélio/virologia , Homeostase/efeitos dos fármacos , Humanos , Proteínas Oncogênicas Virais/antagonistas & inibidores , Proteínas Oncogênicas Virais/metabolismo , Infecções por Papillomavirus/patologia , Infecções por Papillomavirus/virologia , Infecção Persistente/patologia , Infecção Persistente/virologia
13.
Viruses ; 14(2)2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-35215904

RESUMO

The aim of the report was to present the circulation of BVDV (bovine viral diarrhea virus) in the cattle population and determine the cause of the failure of vaccination failure leading to the birth of the PI (persistently infected) calf. The case study was carried out at the BVDV-free animal breeding center and cattle farm, where the vaccination program against BVDV was implemented in 2012, and each newly introduced animal was serologically and virologically tested for BVDV. In this case, a blood sample was taken from a 9-month-old breeding bull. Positive RT-PCR and negative ELISA serology results were obtained. The tests were repeated at 2-week intervals, and the results confirmed the presence of the virus and the absence of specific antibodies, i.e., persistent infection. Additionally, sequencing and phylogenetic analysis were performed, and the BVDV-1d subgenotype was detected. The results of this study showed that pregnant heifers and cows that are vaccinated multiple times with the killed vaccine containing BVDV-1a may not be fully protected against infection with other subgenotypes of BVDV, including their fetuses, which can become PI calves.


Assuntos
Doença das Mucosas por Vírus da Diarreia Viral Bovina/prevenção & controle , Vírus da Diarreia Viral Bovina/imunologia , Doenças Fetais/prevenção & controle , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Animais , Anticorpos Antivirais/sangue , Doença das Mucosas por Vírus da Diarreia Viral Bovina/sangue , Doença das Mucosas por Vírus da Diarreia Viral Bovina/embriologia , Doença das Mucosas por Vírus da Diarreia Viral Bovina/virologia , Bovinos , Vírus da Diarreia Viral Bovina/classificação , Vírus da Diarreia Viral Bovina/genética , Vírus da Diarreia Viral Bovina/isolamento & purificação , Feminino , Doenças Fetais/virologia , Masculino , Infecção Persistente/sangue , Infecção Persistente/virologia , Filogenia , Gravidez , Vacinação , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/genética , Vacinas de Produtos Inativados/imunologia , Vacinas Virais/genética
14.
Cancer Discov ; 12(1): 62-73, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34753749

RESUMO

Coronavirus disease 2019 (COVID-19) infection results in both acute mortality and persistent and/or recurrent disease in patients with hematologic malignancies, but the drivers of persistent infection in this population are unknown. We found that B-cell lymphomas were at particularly high risk for persistent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) positivity. Further analysis of these patients identified discrete risk factors for initial disease severity compared with disease chronicity. Active therapy and diminished T-cell counts were drivers of acute mortality in COVID-19-infected patients with lymphoma. Conversely, B cell-depleting therapy was the primary driver of rehospitalization for COVID-19. In patients with persistent SARS-CoV-2 positivity, we observed high levels of viral entropy consistent with intrahost viral evolution, particularly in patients with impaired CD8+ T-cell immunity. These results suggest that persistent COVID-19 infection is likely to remain a risk in patients with impaired adaptive immunity and that additional therapeutic strategies are needed to enable viral clearance in this high-risk population. SIGNIFICANCE: We describe the largest cohort of persistent symptomatic COVID-19 infection in patients with lymphoid malignancies and identify B-cell depletion as the key immunologic driver of persistent infection. Furthermore, we demonstrate ongoing intrahost viral evolution in patients with persistent COVID-19 infection, particularly in patients with impaired CD8+ T-cell immunity.This article is highlighted in the In This Issue feature, p. 1.


Assuntos
COVID-19/imunologia , COVID-19/virologia , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/virologia , Infecção Persistente/imunologia , Infecção Persistente/virologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Linfócitos B/imunologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Risco , SARS-CoV-2/imunologia , Linfócitos T/imunologia
15.
JCI Insight ; 7(2)2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-34905514

RESUMO

Invariant NK T (iNKT) cells are implicated in viral clearance; however, their role in hepatitis C virus (HCV) infection remains controversial. Here, iNKT cells were studied during different stages of HCV infection. iNKT cells from patients with acute HCV infection and people who inject drugs (PWID) with chronic or spontaneously resolved HCV infection were characterized by flow cytometry. In a longitudinal analysis during acute HCV infection, frequencies of activated CD38+ iNKT cells reproducibly declined in spontaneously resolving patients, whereas they were persistently elevated in patients progressing to chronic infection. During the first year of infection, the frequency of activated CD38+ or CD69+ iNKT cells strongly correlated with alanine transaminase levels with particularly pronounced correlations in spontaneously resolving patients. Increased frequencies of activated iNKT cells in chronic HCV infection were confirmed in cross-sectional analyses of PWID with chronic or spontaneously resolved HCV infection; however, no apparent functional differences were observed with various stimulation protocols. Our data suggest that iNKT cells are activated during acute hepatitis C and that activation is sustained in chronic infection. The correlation between the frequency of activated iNKT cells and alanine transaminase may point toward a role of iNKT cells in liver damage.


Assuntos
ADP-Ribosil Ciclase 1/análise , Antígenos CD/análise , Antígenos de Diferenciação de Linfócitos T/análise , Hepacivirus , Hepatite C , Lectinas Tipo C/análise , Ativação Linfocitária/imunologia , Células T Matadoras Naturais , Doença Aguda , Alanina Transaminase/sangue , Estudos Transversais , Hepacivirus/isolamento & purificação , Hepacivirus/patogenicidade , Hepacivirus/fisiologia , Hepatite C/sangue , Hepatite C/fisiopatologia , Hepatite C/virologia , Humanos , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/virologia , Infecção Persistente/imunologia , Infecção Persistente/virologia , Remissão Espontânea , Carga Viral/imunologia
16.
Pol J Microbiol ; 70(4): 489-500, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34970317

RESUMO

Persistent infection with high-risk human papillomavirus (HR-HPV) is the most important determinate in the development of cervical cancer, and cervical microecology can modulate cervical viral infection. However, few studies have been conducted on the microecological analysis of cervical diseases using strict physiological factors. This study investigated the characteristics and dynamics of cervical microecology in childbearing-age Chinese women with different degrees of HR-HPV-positive cervical lesions. A total of 168 subjects were selected according to the selection criteria, including healthy HPV-negative individuals (n = 29), HR-HPV-infected individuals (n = 29), low-grade squamous intraepithelial lesion individuals (LSIL, n = 32), high-grade squamous intraepithelial lesion individuals (HSIL, n = 40), and cervical cancer individuals (n = 38). We sampled cervical secretions from each subject and performed comparative analysis using the 16S rRNA sequencing method. Comparison analysis showed that Lactobacillus and Ignatzschineria were the dominant genera in the healthy group, while Gardnerella and Prevotella were more enriched in the disease groups. Based on the taxa composition, we roughly divided the development of cervical cancer into two phases: phase I was from healthy status to HR-HPV infection and LSIL; phase II was from LSIL to HSIL and cervical cancer. Different interactions among different genera were observed in different groups. Prevotella inhibited the abundance of Lactobacillus in the healthy group, while Prevotella inhabited the abundance of Gardnerella in the other groups. In the HR-HPV infection group, Ignatzschineria and Enterococcus showed a positive interaction but dissociated with the increase in cervical lesions, which might eventually lead to a continuous decrease in the abundances of Lactobacillus and Ignatzschineria.


Assuntos
Colo do Útero , Infecções por Papillomavirus , Neoplasias do Colo do Útero , Vagina , Adulto , Bactérias/genética , Biodiversidade , Colo do Útero/microbiologia , Colo do Útero/patologia , Colo do Útero/virologia , Feminino , Humanos , Papillomaviridae/genética , Infecções por Papillomavirus/microbiologia , Infecção Persistente/microbiologia , Infecção Persistente/virologia , RNA Ribossômico 16S/genética , Microambiente Tumoral , Neoplasias do Colo do Útero/microbiologia , Neoplasias do Colo do Útero/virologia , Vagina/microbiologia , Vagina/virologia , Adulto Jovem
17.
Ann Clin Microbiol Antimicrob ; 20(1): 85, 2021 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-34969393

RESUMO

BACKGROUND: There is growing evidence that antibody responses play a role in the resolution of SARS-CoV-2 infection. Patients with primary or secondary antibody deficiency are at increased risk of persistent infection. This challenging clinical scenario is associated with adverse patient outcome and potentially creates an ecological niche for the evolution of novel SARS-CoV-2 variants with immune evasion capacity. Case reports and/or series have implied a therapeutic role for convalescent plasma (CP) to secure virological clearance, although concerns have been raised about the effectiveness of CP and its potential to drive viral evolution, and it has largely been withdrawn from clinical use in the UK. CASE PRESENTATION: We report two cases in which persistent SARS-CoV-2 infection was cleared following administration of the monoclonal antibody combination casirivimab and imdevimab (REGN-COV2, Ronapreve). A 55-year-old male with follicular lymphoma, treated with B cell depleting therapy, developed SARS-CoV-2 infection in September 2020 which then persisted for over 200 days. He was hospitalised on four occasions with COVID-19 and suffered debilitating fatigue and malaise throughout. There was no clinical response to antiviral therapy with remdesivir or CP, and SARS-CoV-2 was consistently detected in nasopharyngeal swabs. Intrahost evolution of several spike variants of uncertain significance was identified by viral sequence analysis. Delivery of REGN-COV2, in combination with remdesivir, was associated with clinical improvement and viral clearance within 6 days, which was sustained for over 150 days despite immunotherapy for relapsed follicular lymphoma. The second case, a 68-year-old female with chronic lymphocytic leukaemia on ibrutinib, also developed persistent SARS-CoV-2 infection. Despite a lack of response to remdesivir, infection promptly cleared following REGN-COV2 in combination with remdesivir, accompanied by resolution of inflammation and full clinical recovery that has been maintained for over 290 days. CONCLUSIONS: These cases highlight the potential benefit of REGN-COV2 as therapy for persistent SARS-CoV-2 infection in antibody deficient individuals, including after failure of CP treatment. Formal clinical studies are warranted to assess the effectiveness of REGN-COV2 in antibody-deficient patients, especially in light of the emergence of variants of concern, such as Omicron, that appear to evade REGN-COV2 neutralisation.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Tratamento Farmacológico da COVID-19 , Infecção Persistente/virologia , Idoso , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes , COVID-19/terapia , Combinação de Medicamentos , Feminino , Humanos , Imunização Passiva , Linfoma Folicular , Masculino , Pessoa de Meia-Idade , Infecção Persistente/tratamento farmacológico , SARS-CoV-2 , Resultado do Tratamento , Soroterapia para COVID-19
18.
Proc Natl Acad Sci U S A ; 118(46)2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34753817

RESUMO

Acute HIV-1 infection (AHI) results in the widespread depletion of CD4+ T cells in peripheral blood and gut mucosal tissue. However, the impact on the predominantly CD4+ immunoregulatory invariant natural killer T (iNKT) cells during AHI remains unknown. Here, iNKT cells from peripheral blood and colonic mucosa were investigated during treated and untreated AHI. iNKT cells in blood were activated and rapidly depleted in untreated AHI. At the time of peak HIV-1 viral load, these cells showed the elevated expression of cell death-associated transcripts compared to preinfection. Residual peripheral iNKT cells suffered a diminished responsiveness to in vitro stimulation early into chronic infection. Additionally, HIV-1 DNA, as well as spliced and unspliced viral RNA, were detected in iNKT cells isolated from blood, indicating the active infection of these cells in vivo. The loss of iNKT cells occurred from Fiebig stage III in the colonic mucosa, and these cells were not restored to normal levels after initiation of ART during AHI. CD4+ iNKT cells were depleted faster and more profoundly than conventional CD4+ T cells, and the preferential infection of CD4+ iNKT cells over conventional CD4+ T cells was confirmed by in vitro infection experiments. In vitro data also provided evidence of latent infection in iNKT cells. Strikingly, preinfection levels of peripheral blood CD4+ iNKT cells correlated directly with the peak HIV-1 load. These findings support a model in which iNKT cells are early targets for HIV-1 infection, driving their rapid loss from circulation and colonic mucosa.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Colo/imunologia , Colo/virologia , Infecções por HIV/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/virologia , Células T Matadoras Naturais/imunologia , Adolescente , Adulto , Progressão da Doença , Feminino , Infecções por HIV/virologia , HIV-1/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Infecção Persistente/imunologia , Infecção Persistente/virologia , Adulto Jovem
19.
Viruses ; 13(11)2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34834923

RESUMO

Mosquito-borne viruses of the Flavivirus genus (Flaviviridae family) pose an ongoing threat to global public health. For example, dengue, Japanese encephalitis, West Nile, yellow fever, and Zika viruses are transmitted by infected mosquitoes and cause severe and fatal diseases in humans. The means by which mosquito-borne flaviviruses establish persistent infection in mosquitoes and cause disease in humans are complex and depend upon a myriad of virus-host interactions, such as those of the innate immune system, which are the main focus of our review. This review also covers the different strategies utilized by mosquito-borne flaviviruses to antagonize the innate immune response in humans and mosquitoes. Given the lack of antiviral therapeutics for mosquito-borne flaviviruses, improving our understanding of these virus-immune interactions could lead to new antiviral therapies and strategies for developing refractory vectors incapable of transmitting these viruses, and can also provide insights into determinants of viral tropism that influence virus emergence into new species.


Assuntos
Culicidae/imunologia , Infecções por Flavivirus/imunologia , Infecções por Flavivirus/veterinária , Flavivirus/imunologia , Infecção Persistente/imunologia , Infecção Persistente/veterinária , Animais , Culicidae/fisiologia , Culicidae/virologia , Flavivirus/genética , Flavivirus/fisiologia , Infecções por Flavivirus/transmissão , Infecções por Flavivirus/virologia , Humanos , Imunidade Inata , Mosquitos Vetores/imunologia , Mosquitos Vetores/fisiologia , Mosquitos Vetores/virologia , Infecção Persistente/virologia
20.
Curr Opin Virol ; 51: 106-110, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34628358

RESUMO

The ability to establish long term persistent infection is a feature of human papillomaviruses. The available evidence is that this ability is a consequence of a complex local immune milieu whereby innate immune receptors and signalling pathway cascades are inhibited by HPV early proteins resulting in failure of dendritic cell maturation, antigen processing and presentation and activation of cytotoxic antigen specific T cell responses. The development of cutaneous and mucosal infection models with the mouse papillomavirus MmuPV1 and the access to multiple gene deficient strains is providing the frame work to dissect the mechanisms underlying these complex host virus interactions.


Assuntos
Interações entre Hospedeiro e Microrganismos/imunologia , Evasão da Resposta Imune , Papillomaviridae/imunologia , Papillomaviridae/patogenicidade , Infecções por Papillomavirus/imunologia , Infecções por Papillomavirus/virologia , Infecção Persistente/virologia , Animais , Apresentação de Antígeno , Células Dendríticas , Humanos , Infecção Persistente/imunologia , Linfócitos T
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...