Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Infect Dis ; 223(11): 1842-1854, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33837392

RESUMEN

BACKGROUND: Coronavirus disease 2019 (COVID-19) patients manifest with pulmonary symptoms reflected by diffuse alveolar damage (DAD), excessive inflammation, and thromboembolism. The mechanisms mediating these processes remain unclear. METHODS: We performed multicolor staining for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) proteins and lineage markers to define viral tropism and lung pathobiology in 5 autopsy cases. RESULTS: Lung parenchyma showed severe DAD with thromboemboli. Viral infection was found in an extensive range of cells including pneumocyte type II, ciliated, goblet, club-like, and endothelial cells. More than 90% of infiltrating immune cells were positive for viral proteins including macrophages, monocytes, neutrophils, natural killer (NK) cells, B cells, and T cells. Most but not all infected cells were angiotensin-converting enzyme 2 (ACE2) positive. The numbers of infected and ACE2-positive cells are associated with extensive tissue damage. Infected tissues exhibited high levels of inflammatory cells including macrophages, monocytes, neutrophils, and NK cells, and low levels of B cells but abundant T cells consisting of mainly T helper cells, few cytotoxic T cells, and no regulatory T cells. Robust interleukin-6 expression was present in most cells, with or without infection. CONCLUSIONS: In fatal COVID-19 lungs, there are broad SARS-CoV-2 cell tropisms, extensive infiltrated innate immune cells, and activation and depletion of adaptive immune cells, contributing to severe tissue damage, thromboemboli, excess inflammation, and compromised immune responses.


Asunto(s)
COVID-19/patología , Pulmón/patología , SARS-CoV-2/fisiología , Tropismo Viral , Adulto , Anciano , COVID-19/inmunología , COVID-19/virología , Femenino , Humanos , Inmunidad Innata , Pulmón/citología , Pulmón/inmunología , Pulmón/virología , Masculino , Persona de Mediana Edad , Alveolos Pulmonares/inmunología , Alveolos Pulmonares/patología , Alveolos Pulmonares/virología , Tropismo Viral/inmunología
3.
Sci Rep ; 10(1): 14289, 2020 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-32868837

RESUMEN

Ebola virus (EBOV) is an enveloped, single-stranded RNA virus that can cause Ebola virus disease (EVD). It is thought that EVD survivors are protected against subsequent infection with EBOV and that neutralising antibodies to the viral surface glycoprotein (GP) are potential correlates of protection. Serological studies are vital to assess neutralising antibodies targeted to EBOV GP; however, handling of EBOV is limited to containment level 4 laboratories. Pseudotyped viruses can be used as alternatives to live viruses, which require high levels of bio-containment, in serological and viral entry assays. However, neutralisation capacity can differ among pseudotyped virus platforms. We evaluated the suitability of EBOV GP pseudotyped human immunodeficiency virus type 1 (HIV-1) and vesicular stomatitis virus (VSV) to measure the neutralising ability of plasma from EVD survivors, when compared to results from a live EBOV neutralisation assay. The sensitivity, specificity and correlation with live EBOV neutralisation were greater for the VSV-based pseudotyped virus system, which is particularly important when evaluating EBOV vaccine responses and immuno-therapeutics. Therefore, the EBOV GP pseudotyped VSV neutralisation assay reported here could be used to provide a better understanding of the putative correlates of protection against EBOV.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Ebolavirus/inmunología , VIH-1/inmunología , Vesiculovirus/inmunología , Proteínas del Envoltorio Viral/inmunología , Humanos , Pruebas de Neutralización , Tropismo Viral/inmunología
4.
J Virol ; 94(12)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32295903

RESUMEN

Cell entry by HIV-1 is mediated by its principal receptor, CD4, and a coreceptor, either CCR5 or CXCR4, with viral envelope glycoprotein gp120. Generally, CCR5-using HIV-1 variants, called R5, predominate over most of the course of infection, while CXCR4-using HIV-1 variants (variants that utilize both CCR5 and CXCR4 [R5X4, or dual] or CXCR4 alone [X4]) emerge at late-stage infection in half of HIV-1-infected individuals and are associated with disease progression. Although X4 variants also appear during acute-phase infection in some cases, these variants apparently fall to undetectable levels thereafter. In this study, replication-competent X4 variants were isolated from plasma of drug treatment-naive individuals infected with HIV-1 strain CRF01_AE, which dominantly carries viral RNA (vRNA) of R5 variants. Next-generation sequencing (NGS) confirmed that sequences of X4 variants were indeed present in plasma vRNA from these individuals as a minor population. On the other hand, in one individual with a mixed infection in which X4 variants were dominant, only R5 replication-competent variants were isolated from plasma. These results indicate the existence of replication-competent variants with different coreceptor usage as minor populations.IMPORTANCE The coreceptor switch of HIV-1 from R5 to CXCR4-using variants (R5X4 or X4) has been observed in about half of HIV-1-infected individuals at late-stage infection with loss of CD4 cell count and disease progression. However, the mechanisms that underlie the emergence of CXCR4-using variants at this stage are unclear. In the present study, CXCR4-using X4 variants were isolated from plasma samples of HIV-1-infected individuals that dominantly carried vRNA of R5 variants. The sequences of the X4 variants were detected as a minor population using next-generation sequencing. Taken together, CXCR4-using variants at late-stage infection are likely to emerge when replication-competent CXCR4-using variants are maintained as a minor population during the course of infection. The present study may support the hypothesis that R5-to-X4 switching is mediated by the expansion of preexisting X4 variants in some cases.


Asunto(s)
Infecciones por VIH/inmunología , VIH-1/genética , Receptores CCR5/genética , Receptores CXCR4/genética , Receptores del VIH/inmunología , Adulto , Anciano , Secuencia de Aminoácidos , Recuento de Linfocito CD4 , Coinfección , Progresión de la Enfermedad , Femenino , Regulación de la Expresión Génica , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/clasificación , VIH-1/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Masculino , Persona de Mediana Edad , Filogenia , Unión Proteica , ARN Viral/genética , ARN Viral/inmunología , Receptores CCR5/inmunología , Receptores CXCR4/inmunología , Receptores del VIH/genética , Tropismo Viral/genética , Tropismo Viral/inmunología , Acoplamiento Viral , Internalización del Virus
5.
J Gen Virol ; 101(6): 599-608, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32213247

RESUMEN

Infection of chicken coronavirus infectious bronchitis virus (IBV) is initiated by binding of the viral heavily N-glycosylated attachment protein spike to the alpha-2,3-linked sialic acid receptor Neu5Ac. Previously, we have shown that N-glycosylation of recombinantly expressed receptor binding domain (RBD) of the spike of IBV-M41 is of critical importance for binding to chicken trachea tissue. Here we investigated the role of N-glycosylation of the RBD on receptor specificity and virus replication in the context of the virus particle. Using our reverse genetics system we were able to generate recombinant IBVs for nine-out-of-ten individual N-glycosylation mutants. In vitro growth kinetics of these viruses were comparable to the virus containing the wild-type M41-S1. Furthermore, Neu5Ac binding by the recombinant viruses containing single N-glycosylation site knock-out mutations matched the Neu5Ac binding observed with the recombinant RBDs. Five N-glycosylation mutants lost the ability to bind Neu5Ac and gained binding to a different, yet unknown, sialylated glycan receptor on host cells. These results demonstrate that N-glycosylation of IBV is a determinant for receptor specificity.


Asunto(s)
Infecciones por Coronavirus/inmunología , Especificidad del Huésped/inmunología , Virus de la Bronquitis Infecciosa/química , Dominios Proteicos , Receptores Virales/inmunología , Glicoproteína de la Espiga del Coronavirus/química , Animales , Línea Celular , Embrión de Pollo , Infecciones por Coronavirus/virología , Glicosilación , Virus de la Bronquitis Infecciosa/inmunología , Riñón/citología , Riñón/embriología , Unión Proteica , Receptores de Superficie Celular/metabolismo , Receptores Virales/metabolismo , Proteínas Recombinantes , Glicoproteína de la Espiga del Coronavirus/metabolismo , Tropismo Viral/inmunología , Acoplamiento Viral , Replicación Viral
6.
J Virol ; 94(2)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31666372

RESUMEN

Follicular helper T (TFH) cells have been shown to support productive human immunodeficiency virus type 1 (HIV-1) replication and to serve as a key component of the latent viral reservoir. However, the viral characteristics of this latent reservoir and the clinical relevance of this reservoir remain unclear. In this study, we assessed the tropic composition of latent viruses from peripheral TFH (pTFH), non-TFH memory, and naive CD4+ T cells from individuals with HIV-1 infections on suppressive combined antiretroviral therapy (cART). X4-tropic latent HIV-1 was preferentially enriched in pTFH cells compared to levels in the other two subsets. Interestingly, the ratio of X4-tropic latent HIV-1 in pTFH cells not only was robustly and inversely correlated with blood CD4+ T cell counts across patients but also was prognostic of CD4+ T cell recovery in individuals on long-term cART. Moreover, patients with higher X4-tropic latent HIV-1 ratios in pTFH cells showed greater risks of opportunistic coinfections. These findings reveal the characteristics of latent HIV-1 in TFH cells and suggest that the ratio of X4-tropic latent HIV-1 in pTFH cells is a valuable indicator for disease progression and cART efficacy.IMPORTANCE TFH cells have been shown to harbor a significant amount of latent HIV-1; however, the viral characteristics of this reservoir and its clinical relevance remain largely unknown. In this study, we demonstrate that X4-tropic latent HIV-1 is preferentially enriched in pTFH cells, which also accurately reflects the viral tropism shift. The ratio of X4-tropic proviruses in pTFH cells but not in other memory CD4+ T cell subsets is inversely and closely correlated with blood CD4+ T cell counts and CD4+ T cell recovery rates with cART. Our data suggest that the ratio of X4-tropic provirus in peripheral TFH cells can be easily measured and reflects disease progression and treatment outcomes during cART.


Asunto(s)
Infecciones por VIH , VIH-1/fisiología , Memoria Inmunológica , Provirus/fisiología , Linfocitos T Colaboradores-Inductores , Tropismo Viral/inmunología , Latencia del Virus/inmunología , Adulto , Recuento de Linfocito CD4 , Progresión de la Enfermedad , Femenino , Infecciones por VIH/inmunología , Infecciones por VIH/patología , Humanos , Masculino , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/patología , Linfocitos T Colaboradores-Inductores/virología
7.
J Virol ; 94(4)2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31776276

RESUMEN

Influenza A virus (IAV) is a human respiratory pathogen that causes yearly global epidemics, as well as sporadic pandemics due to human adaptation of pathogenic strains. Efficient replication of IAV in different species is, in part, dictated by its ability to exploit the genetic environment of the host cell. To investigate IAV tropism in human cells, we evaluated the replication of IAV strains in a diverse subset of epithelial cell lines. HeLa cells were refractory to the growth of human H1N1 and H3N2 viruses and low-pathogenic avian influenza (LPAI) viruses. Interestingly, a human isolate of the highly pathogenic avian influenza (HPAI) H5N1 virus successfully propagated in HeLa cells to levels comparable to those in a human lung cell line. Heterokaryon cells generated by fusion of HeLa and permissive cells supported H1N1 virus growth, suggesting the absence of a host factor(s) required for the replication of H1N1, but not H5N1, viruses in HeLa cells. The absence of this factor(s) was mapped to reduced nuclear import, replication, and translation, as well as deficient viral budding. Using reassortant H1N1:H5N1 viruses, we found that the combined introduction of nucleoprotein (NP) and hemagglutinin (HA) from an H5N1 virus was necessary and sufficient to enable H1N1 virus growth. Overall, this study suggests that the absence of one or more cellular factors in HeLa cells results in abortive replication of H1N1, H3N2, and LPAI viruses, which can be circumvented upon the introduction of H5N1 virus NP and HA. Further understanding of the molecular basis of this restriction will provide important insights into the virus-host interactions that underlie IAV pathogenesis and tropism.IMPORTANCE Many zoonotic avian influenza A viruses have successfully crossed the species barrier and caused mild to life-threatening disease in humans. While human-to-human transmission is limited, there is a risk that these zoonotic viruses may acquire adaptive mutations enabling them to propagate efficiently and cause devastating human pandemics. Therefore, it is important to identify viral determinants that provide these viruses with a replicative advantage in human cells. Here, we tested the growth of influenza A virus in a subset of human cell lines and found that abortive replication of H1N1 viruses in HeLa cells can be circumvented upon the introduction of H5N1 virus HA and NP. Overall, this work leverages the genetic diversity of multiple human cell lines to highlight viral determinants that could contribute to H5N1 virus pathogenesis and tropism.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/metabolismo , Tropismo Viral/genética , Células A549 , Animales , Aves , Línea Celular , Perros , Células HEK293 , Células HeLa , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/genética , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Virus de la Influenza A/genética , Virus de la Influenza A/metabolismo , Virus de la Influenza A/patogenicidad , Gripe Aviar/genética , Gripe Aviar/metabolismo , Gripe Humana/genética , Gripe Humana/virología , Células de Riñón Canino Madin Darby , Tropismo Viral/inmunología , Replicación Viral/genética
8.
PLoS Pathog ; 15(10): e1008057, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31671153

RESUMEN

Human astroviruses (HAstV) are understudied positive-strand RNA viruses that cause gastroenteritis mostly in children and the elderly. Three clades of astroviruses, classic, MLB-type and VA-type have been reported in humans. One limitation towards a better understanding of these viruses has been the lack of a physiologically relevant cell culture model that supports growth of all clades of HAstV. Herein, we demonstrate infection of HAstV strains belonging to all three clades in epithelium-only human intestinal enteroids (HIE) isolated from biopsy-derived intestinal crypts. A detailed investigation of infection of VA1, a member of the non-canonical HAstV-VA/HMO clade, showed robust replication in HIE derived from different patients and from different intestinal regions independent of the cellular differentiation status. Flow cytometry and immunofluorescence analysis revealed that VA1 infects several cell types, including intestinal progenitor cells and mature enterocytes, in HIE cultures. RNA profiling of VA1-infected HIE uncovered that the host response to infection is dominated by interferon (IFN)-mediated innate immune responses. A comparison of the antiviral host response in non-transformed HIE and transformed human colon carcinoma Caco-2 cells highlighted significant differences between these cells, including an increased magnitude of the response in HIE. Additional studies confirmed the sensitivity of VA1 to exogenous IFNs, and indicated that the endogenous IFN response of HIE to curtail the growth of strains from all three clades. Genotypic variation in the permissiveness of different HIE lines to HAstV could be overcome by pharmacologic inhibition of JAK/STAT signaling. Collectively, our data identify HIE as a universal infection model for HAstV and an improved model of the intestinal epithelium to investigate enteric virus-host interactions.


Asunto(s)
Infecciones por Astroviridae/inmunología , Infecciones por Astroviridae/veterinaria , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Mamastrovirus/fisiología , Tropismo Viral/genética , Animales , Células CACO-2 , Línea Celular , Chlorocebus aethiops , Enterocitos/virología , Gastroenteritis/virología , Humanos , Inmunidad Innata/inmunología , Interferones/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/virología , Intestino Delgado/citología , Intestino Delgado/virología , Mamastrovirus/genética , Mamastrovirus/inmunología , Células Vero , Tropismo Viral/inmunología
9.
FEBS Lett ; 593(24): 3583-3608, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31769017

RESUMEN

Human adenovirus commonly causes infections of respiratory, gastrointestinal, genitourinary, and ocular surface mucosae. Although most adenovirus eye infections are mild and self-limited, specific viruses within human adenovirus species D are associated with epidemic keratoconjunctivitis (EKC), a severe and highly contagious ocular surface infection, which can lead to chronic and/or recurrent, visually disabling keratitis. In this review, we discuss the links between adenovirus ontogeny, genomics, immune responses, and corneal pathogenesis, for those viruses that cause EKC.


Asunto(s)
Adenovirus Humanos/patogenicidad , Evolución Biológica , Proteínas del Ojo/genética , Interacciones Huésped-Patógeno/genética , Queratitis/genética , Queratoconjuntivitis/genética , Proteínas Virales/genética , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Animales , Conjuntiva/inmunología , Conjuntiva/metabolismo , Conjuntiva/patología , Conjuntiva/virología , Córnea/inmunología , Córnea/metabolismo , Córnea/patología , Córnea/virología , Modelos Animales de Enfermedad , Proteínas del Ojo/inmunología , Regulación de la Expresión Génica , Genómica/métodos , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Queratitis/inmunología , Queratitis/patología , Queratitis/virología , Queratoconjuntivitis/inmunología , Queratoconjuntivitis/patología , Queratoconjuntivitis/virología , Filogenia , Proteínas Virales/inmunología , Tropismo Viral/genética , Tropismo Viral/inmunología
10.
Elife ; 82019 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-31644426

RESUMEN

Numerous challenges have impeded HIV-1 vaccine development. Among these is the lack of a convenient small animal model in which to study antibody elicitation and efficacy. We describe a chimeric Rhabdo-Immunodeficiency virus (RhIV) murine model that recapitulates key features of HIV-1 entry, tropism and antibody sensitivity. RhIVs are based on vesicular stomatitis viruses (VSV), but viral entry is mediated by HIV-1 Env proteins from diverse HIV-1 strains. RhIV infection of transgenic mice expressing human CD4 and CCR5, exclusively on mouse CD4+ cells, at levels mimicking those on human CD4+ T-cells, resulted in acute, resolving viremia and CD4+ T-cell depletion. RhIV infection elicited protective immunity, and antibodies to HIV-1 Env that were primarily non-neutralizing and had modest protective efficacy following passive transfer. The RhIV model enables the convenient in vivo study of HIV-1 Env-receptor interactions, antiviral activity of antibodies and humoral responses against HIV-1 Env, in a genetically manipulatable host.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Linfocitos T CD4-Positivos/inmunología , VIH-1/genética , Virus Reordenados/genética , Vesiculovirus/genética , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética , Animales , Especificidad de Anticuerpos , Antígenos CD4/genética , Antígenos CD4/inmunología , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/virología , Modelos Animales de Enfermedad , Efecto Fundador , Expresión Génica , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/inmunología , Humanos , Ratones , Ratones Transgénicos , Virus Reordenados/inmunología , Receptores CCR5/genética , Receptores CCR5/inmunología , Vesiculovirus/inmunología , Tropismo Viral/genética , Tropismo Viral/inmunología , Internalización del Virus , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología
11.
Viruses ; 11(9)2019 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-31540120

RESUMEN

Old world hantaviruses cause hemorrhagic fever with renal syndrome (HFRS) upon zoonotic transmission to humans. In Europe, the Puumala virus (PUUV) is the main causative agent of HFRS. Tula virus (TULV) is also widely distributed in Europe, but there is little knowledge about the pathogenicity of TULV for humans, as reported cases are rare. We studied the replication of TULV in different cell types in comparison to the pathogenic PUUV and analyzed differences in stimulation of innate immunity. While both viruses replicated to a similar extent in interferon (IFN)-deficient Vero E6 cells, TULV replication in human lung epithelial (A549) cells was slower and less efficient when compared to PUUV. In contrast to PUUV, no replication of TULV could be detected in human microvascular endothelial cells and in macrophages. While a strong innate immune response towards PUUV infection was evident at 48 h post infection, TULV infection triggered only a weak IFN response late after infection of A549 cells. Using appropriate in vitro cell culture models for the orthohantavirus infection, we could demonstrate major differences in host cell tropism, replication kinetics, and innate immune induction between pathogenic PUUV and the presumably non- or low-pathogenic TULV that are not observed in Vero E6 cells and may contribute to differences in virulence.


Asunto(s)
Células Endoteliales/virología , Inmunidad Innata , Macrófagos/virología , Orthohantavirus/inmunología , Virus Puumala/inmunología , Replicación Viral , Células A549 , Animales , Chlorocebus aethiops , Células Endoteliales/inmunología , Orthohantavirus/fisiología , Fiebre Hemorrágica con Síndrome Renal/virología , Humanos , Interferón Tipo I/inmunología , Cinética , Macrófagos/inmunología , Virus Puumala/patogenicidad , Virus Puumala/fisiología , ARN Viral/análisis , Células THP-1 , Células Vero , Tropismo Viral/inmunología , Virulencia/inmunología
12.
Acta Biomater ; 97: 93-104, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31386928

RESUMEN

Adenovirus (Ad) is a promising viral carrier in gene therapy because of its unique attribution. However, clinical applications of Ad vectors are currently restricted by their immunogenicity and broad native tropism. To address these obstacles, a variety of nonimmunogenic polymers are utilized to modify Ad vectors chemically or physically. In this review, we systemically discuss the functions of polymers in Ad-mediated gene delivery from two aspects: evading the host immune responses to Ads and redirecting Ad tropism. With polyethylene glycol (PEG) first in order, a variety of polymers have been developed to shield the surface of Ad vectors and well accomplished to evade the host immune response, block CAR-dependant cellular uptake, and reduce accumulation in the liver. In addition, shielding Ad vectors with targeted polymers (including targeting ligand-conjugated polymers and bio-responsive polymers) can also efficiently retarget Ad vectors to tumor tissues and reduce their distribution in nontargeted tissues. With its potential to evade the immune response and retarget Ad vectors, modification with polymers has been generally regarded as a promising strategy to facilitate the clinical applications of Ad vectors for virotherapy. STATEMENT OF SIGNIFICANCE: There is no doubt that Adenovirus (Ads) are attractive vectors for gene therapy, with high sophistication and effectiveness in overcoming both extra- and intracellular barriers, which cannot be exceeded by any other nonviral gene vectors. Unfortunately, their clinical applications are still restricted by some critical hurdles, including immunogenicity and native broad tropism. Therefore, a variety of elegant strategies have been developed from various angles to address these hurdles. Among these various strategies, coating Ads with nonimmunogenic polymers has attracted much attention. In this review, we systemically discuss the functions of polymers in Ad-mediated gene delivery from two aspects: evading the host immune responses to Ads and redirecting Ad tropism. In addition, the key factors in Ad modification with polymers have been highlighted and summarized to provide guiding theory for the design of more effective and safer polymer-Ad hybrid gene vectors.


Asunto(s)
Adenoviridae , Terapia Genética , Vectores Genéticos , Evasión Inmune/efectos de los fármacos , Polietilenglicoles/uso terapéutico , Transducción Genética , Tropismo Viral , Animales , Vectores Genéticos/inmunología , Vectores Genéticos/uso terapéutico , Humanos , Tropismo Viral/efectos de los fármacos , Tropismo Viral/inmunología
13.
PLoS Pathog ; 15(6): e1007790, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31194854

RESUMEN

Bourbon virus (BRBV) is an emerging tick-borne RNA virus in the orthomyxoviridae family that was discovered in 2014. Although fatal human cases of BRBV have been described, little is known about its pathogenesis, and no antiviral therapies or vaccines exist. We obtained serum from a fatal case in 2017 and successfully recovered the second human infectious isolate of BRBV. Next-generation sequencing of the St. Louis isolate of BRBV (BRBV-STL) showed >99% nucleotide identity to the original reference isolate. Using BRBV-STL, we developed a small animal model to study BRBV-STL tropism in vivo and evaluated the prophylactic and therapeutic efficacy of the experimental antiviral drug favipiravir against BRBV-induced disease. Infection of Ifnar1-/- mice lacking the type I interferon receptor, but not congenic wild-type animals, resulted in uniformly fatal disease 6 to 10 days after infection. RNA in situ hybridization and viral yield assays demonstrated a broad tropism of BRBV-STL with highest levels detected in liver and spleen. In vitro replication and polymerase activity of BRBV-STL were inhibited by favipiravir. Moreover, administration of favipiravir as a prophylaxis or as post-exposure therapy three days after infection prevented BRBV-STL-induced mortality in immunocompromised Ifnar1-/- mice. These results suggest that favipiravir may be a candidate treatment for humans who become infected with BRBV.


Asunto(s)
Amidas/farmacología , Antivirales/farmacología , Infecciones por Orthomyxoviridae/prevención & control , Pirazinas/farmacología , Thogotovirus/inmunología , Animales , Chlorocebus aethiops , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/patología , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/inmunología , Thogotovirus/patogenicidad , Células Vero , Tropismo Viral/efectos de los fármacos , Tropismo Viral/genética , Tropismo Viral/inmunología
14.
Microb Pathog ; 135: 103581, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31175971

RESUMEN

Chikungunya virus (CHIKV) is mosquito-borne alphavirus that has caused epidemics around the world. Many individuals affected by the disease may experience joint pain that persists for months after the acute phase. The pathophysiology of viral arthritis is not completely elucidated. And it is important to emphasize that the effects of the viral infection in each host may depend on host factors that include immune response, as well as factors specific to the virus as tissue tropism. The main pathway for the response against viral infection is through induction of type I interferon (IFN-I), whose function is important to control viral replication. Beside this, T cell and macrophage mediated immunopathology in CHIKV infections has been reported. It has been demonstrated that some association with the Arginase I and macrophages type II are involved in the infection profile along with myeloid-derived suppressor cells (MDSC) that are responsible for T cell suppression. Therefore, in this review, will be discuss an overview on CHIKV immunopathogenesis and the importance of Arginase I.


Asunto(s)
Arginasa/metabolismo , Fiebre Chikungunya/inmunología , Virus Chikungunya/patogenicidad , Interacciones Huésped-Patógeno/inmunología , Citocinas/metabolismo , Humanos , Interferón Tipo I , Macrófagos , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Factores Supresores Inmunológicos , Linfocitos T , Tropismo Viral/inmunología , Replicación Viral
15.
Poult Sci ; 98(9): 3514-3522, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-30993349

RESUMEN

Fowl adenovirus serotype 4 (FAdV-4) is the causative agent of hydropericardium syndrome. To clarify the effects of FAdV-4 on immune organs in birds, we conducted a detailed examination of dynamic morphology and damage mechanisms in chickens randomly divided into 4 groups (FAdV-4, vaccination, FAdV-4 plus vaccination, and control). FAdV-4 caused the depletion of lymphocytes and subsequent growth impairment in the thymus and bursa. Chickens infected with FAdV-4 and subjected to vaccination experienced greater inhibition of antibody responses to inactivated vaccines against Newcastle disease and avian influenza virus subtype H9 than uninfected and vaccinated chickens. The mechanisms underlying adenovirus-mediated lymphoid organ damage were further investigated via transferase-mediated dUTP nick-end labeling and apoptotic genes transcription analyses. Notably, lymphocytes apoptosis in lymphoid organs and expression of specific gene transcripts was significantly upregulated after infection (P < 0.05). Furthermore, increased expression of interleukin (IL)-6, IL-8, and tumor necrosis factor (TNF)-α mRNA was observed (P < 0.05), compared to the control group. Our collective findings suggested that FAdV-4 caused structural and functional damage of immune organs via apoptosis along with induction of a severe inflammatory response.


Asunto(s)
Infecciones por Adenoviridae/veterinaria , Pollos , Adenovirus A Aviar/fisiología , Tolerancia Inmunológica/inmunología , Inmunidad Humoral/inmunología , Enfermedades de las Aves de Corral/inmunología , Tropismo Viral/inmunología , Infecciones por Adenoviridae/inmunología , Animales , Apoptosis , Adenovirus A Aviar/inmunología , Inflamación , Distribución Aleatoria , Serogrupo
16.
PLoS One ; 14(2): e0212882, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30818365

RESUMEN

BACKGROUND: The association between X4 virus and an increased risk of non-AIDS-events has been reported. Morbidity/mortality due to non-AIDS events, which are properly predicted by the CD4/CD8 ratio and VACS index, have become particularly remarkable in HIV-infected patients receiving effective combined antiretroviral therapy (cART). METHODS: We verified the validity of the syllogism: as HIV-tropism (CRT) contributes to the onset of non-AIDS events which are successfully predicted by the CD4/CD8 ratio and VACS index, then CRT correlates with these two variables. The CD4/CD8 ratio and VACS index at baseline and overtime were analyzed according to CRT tested before the first successful cART regimen in newly-diagnosed patients. RESULTS: Patients with R5 variants had a significantly lower baseline VACS percentage risk [mean (95%CI):18.2%(16.1-20.3) vs 24.3%(18.2-22.5), p = 0.002] and higher baseline CD4/CD8 ratio [mean (95%CI):0.43 (0.38-0.47) vs 0.28 (0.19-0.36), p = 0.002] than non-R5 patients. After an initial drop, VACS increased again in R5 and non-R5 patients and the two trend curves almost overlapped. The CD4/CD8 ratio had an increasing trend in both R5 and non-R5 patients; however, even though non-R5 patients had a greater gain of CD4+, they maintained a lower CD4/CD8 ratio at any time point. CONCLUSION: Our study confirms an association between pre-therapy CRT, CD4/CD8 ratio and VACS. A successful cART regimen positively affects the CD4/CD8 ratio; however, the disadvantage conferred by a non-R5 CRT is maintained overtime. The restoration of VACS in all patients could be directly due to variables included in the VACS calculation and to factors that adversely influence these variables.


Asunto(s)
Infecciones por VIH/inmunología , VIH-1 , Adulto , Terapia Antirretroviral Altamente Activa , Recuento de Linfocito CD4 , Relación CD4-CD8 , Estudios de Cohortes , Femenino , Variación Genética , Infecciones por VIH/complicaciones , Infecciones por VIH/mortalidad , VIH-1/genética , VIH-1/fisiología , Humanos , Italia/epidemiología , Masculino , Persona de Mediana Edad , Receptores CCR5/genética , Receptores CXCR4/genética , Factores de Riesgo , Veteranos , Tropismo Viral/genética , Tropismo Viral/inmunología
17.
Nature ; 567(7746): 109-112, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30787439

RESUMEN

Zoonotic influenza A viruses of avian origin can cause severe disease in individuals, or even global pandemics, and thus pose a threat to human populations. Waterfowl and shorebirds are believed to be the reservoir for all influenza A viruses, but this has recently been challenged by the identification of novel influenza A viruses in bats1,2. The major bat influenza A virus envelope glycoprotein, haemagglutinin, does not bind the canonical influenza A virus receptor, sialic acid or any other glycan1,3,4, despite its high sequence and structural homology with conventional haemagglutinins. This functionally uncharacterized plasticity of the bat influenza A virus haemagglutinin means the tropism and zoonotic potential of these viruses has not been fully determined. Here we show, using transcriptomic profiling of susceptible versus non-susceptible cells in combination with genome-wide CRISPR-Cas9 screening, that the major histocompatibility complex class II (MHC-II) human leukocyte antigen DR isotype (HLA-DR) is an essential entry determinant for bat influenza A viruses. Genetic ablation of the HLA-DR α-chain rendered cells resistant to infection by bat influenza A virus, whereas ectopic expression of the HLA-DR complex in non-susceptible cells conferred susceptibility. Expression of MHC-II from different bat species, pigs, mice or chickens also conferred susceptibility to infection. Notably, the infection of mice with bat influenza A virus resulted in robust virus replication in the upper respiratory tract, whereas mice deficient for MHC-II were resistant. Collectively, our data identify MHC-II as a crucial entry mediator for bat influenza A viruses in multiple species, which permits a broad vertebrate tropism.


Asunto(s)
Quirópteros/virología , Antígenos de Histocompatibilidad Clase II/metabolismo , Especificidad del Huésped , Virus de la Influenza A/inmunología , Virus de la Influenza A/fisiología , Zoonosis/inmunología , Zoonosis/virología , Animales , Proteína 9 Asociada a CRISPR , Sistemas CRISPR-Cas , Pollos/genética , Pollos/inmunología , Quirópteros/genética , Quirópteros/inmunología , Quirópteros/metabolismo , Femenino , Perfilación de la Expresión Génica , Antígenos HLA-DR/genética , Antígenos HLA-DR/inmunología , Antígenos HLA-DR/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Especificidad del Huésped/genética , Especificidad del Huésped/inmunología , Humanos , Masculino , Ratones , Ratones Noqueados , Sistema Respiratorio/virología , Porcinos/genética , Porcinos/inmunología , Tropismo Viral/genética , Tropismo Viral/inmunología , Replicación Viral , Zoonosis/genética , Zoonosis/metabolismo
18.
Science ; 360(6385): 204-208, 2018 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-29650672

RESUMEN

Complex interactions between host immunity and the microbiome regulate norovirus infection. However, the mechanism of host immune promotion of enteric virus infection remains obscure. The cellular tropism of noroviruses is also unknown. Recently, we identified CD300lf as a murine norovirus (MNoV) receptor. In this study, we have shown that tuft cells, a rare type of intestinal epithelial cell, express CD300lf and are the target cell for MNoV in the mouse intestine. We found that type 2 cytokines, which induce tuft cell proliferation, promote MNoV infection in vivo. These cytokines can replace the effect of commensal microbiota in promoting virus infection. Our work thus provides insight into how the immune system and microbes can coordinately promote enteric viral infection.


Asunto(s)
Infecciones por Caliciviridae/inmunología , Enterocitos/inmunología , Enterocitos/virología , Microbiota/inmunología , Norovirus/fisiología , Tropismo Viral/inmunología , Animales , Proliferación Celular , Citocinas/metabolismo , Ratones , Receptores Inmunológicos/metabolismo
19.
Adv Virus Res ; 100: 247-278, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29551139

RESUMEN

The use of Drosophila as a model organism has made an important contribution to our understanding of the function and regulation of innate immunity in insects. Indeed, insects can discriminate between different types of pathogens and mount specific and effective responses. Strikingly, the same pathogen can trigger a different immune response in the same organism, depending solely on the route of infection by which the pathogen is delivered. In this review, we recapitulate what is known about antiviral responses in Drosophila, and how they are triggered depending on the route and the mode used for the virus to infect its host.


Asunto(s)
Dicistroviridae/fisiología , Drosophila melanogaster/inmunología , Drosophila melanogaster/virología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/inmunología , Tropismo Viral/inmunología , Animales , Dicistroviridae/inmunología , Modelos Animales de Enfermedad , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Interferencia de ARN/inmunología , Transducción de Señal
20.
Virus Res ; 244: 99-109, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29141204

RESUMEN

In this study, we isolated an infectious bronchitis virus, designated I1101/16, from broiler breeders in China. Analysis of the S1 gene showed that isolate I1101/16 was genetically close to strain ck/CH/LJL/140901, which belongs to the TW I genotype (also known as lineage GI-7 based on the recent IBV classification), however the S2 gene showed genetic diversity comparing to that of S1 gene. Comparison of the genomic sequences showed that the genome of isolate I1101/16 was similar to that of strain ck/CH/LJL/140901 from the 5' end of the genome to the 5' end of the S2 gene and from the 5' end of the 3a gene to the end of the genome, whereas the remaining parts of the genome sequences were more closely related to those of strain 4/91 than those of ck/CH/LJL/140901, thereby suggesting that recombination might have occurred during the origin of the virus. SimPlot and Bootscan analysis of the complete genomic sequence confirmed this hypothesis, where it showed that isolate I1101/16 arose through recombination events between ck/CH/LJL/140901- and 4/91-like viruses. Isolate I1101/16 and strain ck/CH/LJL/140901 shared identical amino acids in almost all five of their B cell epitopes, but the two viruses had a serotype relatedness value of 65, which is well below 80, i.e., the lower cutoff value for viruses of the same serotype. In addition, pathogenicity tests demonstrated that isolate I1101/16 was more pathogenic to 1-day-old specific-pathogen-free chickens than strain ck/CH/LJL/140901, according to analysis of the clinical signs, whereas strain ck/CH/LJL/140901 exhibited prolonged replication and shedding after challenge compared with isolate I1101/16. This study did not provide evidence that recombination can directly alter the antigenicity, virulence, replication, shedding, and tissue tropism of a virus, but it did show that recombination events are likely to be major determinants of viral evolution.


Asunto(s)
Antígenos Virales/genética , Infecciones por Coronavirus/veterinaria , Genoma Viral , Virus de la Bronquitis Infecciosa/genética , Enfermedades de las Aves de Corral/virología , Virus Reordenados/genética , Animales , Antígenos Virales/inmunología , Secuencia de Bases , Embrión de Pollo , Pollos , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Genotipo , Virus de la Bronquitis Infecciosa/crecimiento & desarrollo , Virus de la Bronquitis Infecciosa/patogenicidad , Riñón/inmunología , Riñón/patología , Riñón/virología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/patología , Virus Reordenados/crecimiento & desarrollo , Virus Reordenados/patogenicidad , Alineación de Secuencia , Homología de Secuencia de Ácido Nucleico , Serogrupo , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Tráquea/inmunología , Tráquea/patología , Tráquea/virología , Tropismo Viral/genética , Tropismo Viral/inmunología , Virulencia , Esparcimiento de Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...