Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.396
Filtrar
1.
J Virol ; 97(4): e0167022, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-36971588

RESUMEN

Elucidating the mechanisms underlying the persistence and location of the HIV reservoir is critical for developing cure interventions. While it has been shown that levels of T-cell activation and the size of the HIV reservoir are greater in rectal tissue and lymph nodes (LN) than in blood, the relative contributions of T-cell subsets to this anatomic difference are unknown. We measured and compared HIV-1 DNA content, expression of the T-cell activation markers CD38 and HLA-DR, and expression of the exhaustion markers programmed cell death protein 1 (PD-1) and T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) in naive, central memory (CM), transitional memory (TM), and effector memory (EM) CD4+ and CD8+ T-cells in paired blood and LN samples among 14 people with HIV who were receiving antiretroviral therapy. HIV-1 DNA levels, T-cell immune activation, and TIGIT expression were higher in LN than in blood, especially in CM and TM CD4+ T-cell subsets. Immune activation was significantly higher in all CD8+ T-cell subsets, and memory CD8+ T-cell subsets from LN had higher levels of PD-1 expression, compared with blood, while TIGIT expression levels were significantly lower in TM CD8+ T-cells. The differences seen in CM and TM CD4+ T-cell subsets were more pronounced among participants with CD4+ T-cell counts of <500 cells/µL within 2 years after antiretroviral therapy initiation, thus highlighting increased residual dysregulation in LN as a distinguishing feature of and a potential mechanism for individuals with suboptimal CD4+ T-cell recovery during antiretroviral therapy. IMPORTANCE This study provides new insights into the contributions of different CD4+ and CD8+ T-cell subsets to the anatomic differences between LN and blood in individuals with HIV who have optimal versus suboptimal CD4+ T-cell recovery. To our knowledge, this is the first study comparing paired LN and blood CD4+ and CD8+ T-cell differentiation subsets, as well as those subsets in immunological responders versus immunological suboptimal responders.


Asunto(s)
Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , ADN Viral , Infecciones por VIH , Ganglios Linfáticos , Activación de Linfocitos , Humanos , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/virología , ADN Viral/análisis , VIH-1 , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/virología , Sangre/inmunología , Sangre/virología , Activación de Linfocitos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Masculino , Adulto , Persona de Mediana Edad , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/virología
2.
J Infect Dis ; 225(12): 2167-2175, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35275599

RESUMEN

Starting antiretroviral therapy (ART) in Fiebig 1 acute HIV infection limits the size of viral reservoirs in lymphoid tissues, but does not impact time to virus rebound during a treatment interruption. To better understand why the reduced reservoir size did not increase the time to rebound we measured the frequency and location of HIV RNA+ cells in lymph nodes from participants in the RV254 acute infection cohort. HIV RNA+ cells were detected more frequently and in greater numbers when ART was initiated in Fiebig 1 compared to later Fiebig stages and were localized to the T-cell zone compared to the B-cell follicle with treatment in later Fiebig stages. Variability of virus production in people treated during acute infection suggests that the balance between virus-producing cells and the immune response to clear infected cells rapidly evolves during the earliest stages of infection. Clinical Trials Registration: NCT02919306.


Asunto(s)
Infecciones por VIH , Ganglios Linfáticos , ARN Viral , Antirretrovirales/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/patología , Humanos , Ganglios Linfáticos/virología , ARN Viral/aislamiento & purificación
3.
Viruses ; 14(1)2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-35062287

RESUMEN

African swine fever (ASF) has spread across the globe and has reached closer to North America since being reported in the Dominican Republic and Haiti. As a result, surveillance measures have been heightened and the utility of alternative samples for herd-level monitoring and dead pig sampling have been investigated. Passive surveillance based on the investigation of dead pigs, both domestic and wild, plays a pivotal role in the early detection of an ASF incursion. The World Organization for Animal Health (OIE)-recommended samples for dead pigs are spleen, lymph nodes, bone marrow, lung, tonsil and kidney. However, obtaining these samples requires opening up the carcasses, which is time-consuming, requires skilled labour and often leads to contamination of the premises. As a result, we investigated the suitability of superficial inguinal lymph nodes (SILNs) for surveillance of dead animals. SILNs can be collected in minutes with no to minimum environmental contamination. Here, we demonstrate that the ASF virus (ASFV) genome copy numbers in SILNs highly correlate with those in the spleen and, by sampling SILN, we can detect all pigs that succumb to highly virulent and moderately virulent ASFV strains (100% sensitivity). ASFV was isolated from all positive SILN samples. Thus, sampling SILNs could be useful for routine surveillance of dead pigs on commercial and backyard farms, holding pens and dead on arrival at slaughter houses, as well as during massive die-offs of pigs due to unknown causes.


Asunto(s)
Virus de la Fiebre Porcina Africana/aislamiento & purificación , Fiebre Porcina Africana/diagnóstico , Ganglios Linfáticos/virología , Fiebre Porcina Africana/epidemiología , Fiebre Porcina Africana/virología , Virus de la Fiebre Porcina Africana/genética , Animales , Monitoreo Epidemiológico , Genoma Viral , Bazo/virología , Porcinos
4.
Sci Rep ; 12(1): 135, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34997007

RESUMEN

Interleukin-7 (IL-7) is a cytokine known for its importance in T cell development and survival. How IL-7 shapes CD8 T cell responses during an acute viral infection is less understood. We had previously shown that IL-7 signaling deficient mice have reduced accumulation of influenza-specific CD8 T cells following influenza infection. We sought to determine whether IL-7 affects early CD8 T cell expansion in the mediastinal lymph node and effector function in the lungs. Using IL-7Rα signaling deficient mice, we show that IL-7 is required for a normal sized mediastinal lymph node and the early clonal expansion of influenza-specific CD8 T cells therein. We show that IL-7 plays a cell-intrinsic role in the accumulation of NP366-374 and PA224-233-specific CD8 T cells in the lymph node. We also found that IL-7 shapes terminal differentiation, degranulation and cytokine production to a greater extent in PA224-233-specific than NP366-374-specific CD8 T cells. We further demonstrate that IL-7 is induced in the lung tissue by viral infection and we characterize multiple cellular sources that contribute to IL-7 production. Our findings on IL-7 and its effects on lower respiratory diseases will be important for expanding the utility of therapeutics that are currently available.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Interleucina-7/metabolismo , Pulmón/metabolismo , Ganglios Linfáticos/metabolismo , Infecciones por Orthomyxoviridae/metabolismo , Células A549 , Animales , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/virología , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Patógeno , Humanos , /patogenicidad , Interleucina-7/genética , Subunidad alfa del Receptor de Interleucina-7/genética , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Pulmón/inmunología , Pulmón/virología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/virología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Transducción de Señal
5.
Virology ; 567: 77-86, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35032866

RESUMEN

Type-I interferon (IFN-I) signals exert a critical role in disease progression during viral infections. However, the immunomodulatory mechanisms by which IFN-I dictates disease outcomes remain to be fully defined. Here we report that IFN-I signals mediate thymic atrophy in viral infections, with more severe and prolonged loss of thymic output and unique kinetics and subtypes of IFN-α/ß expression in chronic infection compared to acute infection. Loss of thymic output was linked to inhibition of early stages of thymopoiesis (DN1-DN2 transition, and DN3 proliferation) and pronounced apoptosis during the late DP stage. Notably, infection-associated thymic defects were largely abrogated upon ablation of IFNαßR and partially mitigated in the absence of CD8 T cells, thus implicating direct as well as indirect effects of IFN-I on thymocytes. These findings provide mechanistic underpinnings for immunotherapeutic strategies targeting IFN-1 signals to manipulate disease outcomes during chronic infections and cancers.


Asunto(s)
Atrofia/virología , Interferón-alfa/inmunología , Interferón beta/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Timocitos/virología , Timo/virología , Animales , Atrofia/genética , Atrofia/inmunología , Atrofia/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Enfermedad Crónica , Femenino , Regulación de la Expresión Génica , Humanos , Memoria Inmunológica , Interferón-alfa/genética , Interferón beta/genética , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ganglios Linfáticos/virología , Depleción Linfocítica , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Transducción de Señal/inmunología , Análisis de la Célula Individual , Timocitos/inmunología , Timocitos/patología , Timo/inmunología , Timo/patología
6.
J Virol ; 96(4): e0186721, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34878888

RESUMEN

Common to all cytomegalovirus (CMV) genomes analyzed to date is the presence of G protein-coupled receptors (GPCR). Animal models of CMV provide insights into their role in viral fitness. The mouse cytomegalovirus (MCMV) GPCR, M33, facilitates dendritic cell (DC)-dependent viremia, the extravasation of blood-borne infected DCs to the salivary gland, and the frequency of reactivation events from latently infected tissue explants. Constitutive G protein-coupled M33 signaling is required for these phenotypes, although the contribution of distinct biochemical pathways activated by M33 is unknown. M33 engages Gq/11 to constitutively activate phospholipase C ß (PLCß) and downstream cyclic AMP response-element binding protein (CREB) in vitro. Identification of a MCMV M33 mutant (M33ΔC38) for which CREB signaling was disabled but PLCß activation was preserved provided the opportunity to investigate their relevance in vivo. Following intranasal infection with MCMV M33ΔC38, the absence of M33 CREB Gq/11-dependent signaling correlated with reduced mobilization of lytically-infected DCs to the draining lymph node high endothelial venules (HEVs) and reduced viremia compared with wild type MCMV. In contrast, M33ΔC38-infected DCs within the vascular compartment extravasated to the salivary glands via a pertussis toxin-sensitive, Gi/o-dependent, and CREB-independent mechanism. In the context of MCMV latency, spleen explants from M33ΔC38-infected mice were markedly attenuated for reactivation. Taken together, these data demonstrate that key features of the MCMV life cycle are coordinated in diverse tissues by distinct pathways of the M33 signaling repertoire. IMPORTANCE G protein-coupled receptors (GPCRs) act as cell surface molecular "switches" that regulate the cellular response to environmental stimuli. All cytomegalovirus (CMV) genomes analyzed to date possess GPCR homologs with phylogenetic evidence for independent gene capture events, signifying important in vivo roles. The mouse CMV (MCMV) GPCR homolog, designated M33, is important for cell-associated virus spread and the establishment and/or reactivation of latent MCMV infection. The signaling repertoire of M33 is distinct from cellular GPCRs and little is known of the relevance of component signaling pathways for in vivo M33 function. In this report, we showed that temporal and tissue-specific M33 signaling was required to facilitate in vivo infection. Understanding the relevance of the viral GPCR signaling profiles for in vivo function will provide opportunities for future targeted interventions.


Asunto(s)
Infecciones por Herpesviridae/virología , Muromegalovirus/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Virales/metabolismo , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Células Dendríticas/virología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Infecciones por Herpesviridae/metabolismo , Ganglios Linfáticos/virología , Ratones , Ratones Endogámicos BALB C , Muromegalovirus/genética , Muromegalovirus/metabolismo , Mutación , Fosfolipasa C beta/metabolismo , Receptores Acoplados a Proteínas G/genética , Glándulas Salivales/virología , Transducción de Señal , Proteínas Virales/genética , Viremia/metabolismo , Viremia/virología , Activación Viral/genética
7.
J Immunol Methods ; 501: 113198, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34863818

RESUMEN

The main barrier to a cure for HIV is the persistence of long-lived and proliferating latently infected CD4+ T-cells despite antiretroviral therapy (ART). Latency is well characterized in multiple CD4+ T-cell subsets, however, the contribution of regulatory T-cells (Tregs) expressing FoxP3 as well as immune checkpoints (ICs) PD-1 and CTLA-4 as targets for productive and latent HIV infection in people living with HIV on suppressive ART is less well defined. We used multiplex detection of HIV DNA and RNA with immunohistochemistry (mIHC) on formalin-fixed paraffin embedded (FFPE) cells to simultaneously detect HIV RNA and DNA and cellular markers. HIV DNA and RNA were detected by in situ hybridization (ISH) (RNA/DNAscope) and IHC was used to detect cellular markers (CD4, PD-1, FoxP3, and CTLA-4) by incorporating the tyramide system amplification (TSA) system. We evaluated latently infected cell lines, a primary cell model of HIV latency and excisional lymph node (LN) biopsies collected from people living with HIV (PLWH) on and off ART. We clearly detected infected cells that coexpressed HIV RNA and DNA (active replication) and DNA only (latently infected cells) in combination with IHC markers in the in vitro infection model as well as LN tissue from PLWH both on and off ART. Combining ISH targeting HIV RNA and DNA with IHC provides a platform to detect and quantify HIV persistence within cells identified by multiple markers in tissue samples from PLWH on ART or to study HIV latency.


Asunto(s)
ADN Viral/análisis , Infecciones por VIH/diagnóstico , VIH/genética , Inhibidores de Puntos de Control Inmunológico/análisis , Inmunohistoquímica , Hibridación in Situ , Infección Latente/diagnóstico , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/virología , ARN Viral/análisis , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Células Jurkat , Infección Latente/inmunología , Infección Latente/virología , Valor Predictivo de las Pruebas , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/virología
8.
PLoS Pathog ; 17(12): e1009678, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34855915

RESUMEN

Kyasanur Forest disease virus (KFDV) and the closely related Alkhurma hemorrhagic disease virus (AHFV) are emerging flaviviruses that cause severe viral hemorrhagic fevers in humans. Increasing geographical expansion and case numbers, particularly of KFDV in southwest India, class these viruses as a public health threat. Viral pathogenesis is not well understood and additional vaccines and antivirals are needed to effectively counter the impact of these viruses. However, current animal models of KFDV pathogenesis do not accurately reproduce viral tissue tropism or clinical outcomes observed in humans. Here, we show that pigtailed macaques (Macaca nemestrina) infected with KFDV or AHFV develop viremia that peaks 2 to 4 days following inoculation. Over the course of infection, animals developed lymphocytopenia, thrombocytopenia, and elevated liver enzymes. Infected animals exhibited hallmark signs of human disease characterized by a flushed appearance, piloerection, dehydration, loss of appetite, weakness, and hemorrhagic signs including epistaxis. Virus was commonly present in the gastrointestinal tract, consistent with human disease caused by KFDV and AHFV where gastrointestinal symptoms (hemorrhage, vomiting, diarrhea) are common. Importantly, RNAseq of whole blood revealed that KFDV downregulated gene expression of key clotting factors that was not observed during AHFV infection, consistent with increased severity of KFDV disease observed in this model. This work characterizes a nonhuman primate model for KFDV and AHFV that closely resembles human disease for further utilization in understanding host immunity and development of antiviral countermeasures.


Asunto(s)
Modelos Animales de Enfermedad , Virus de la Encefalitis Transmitidos por Garrapatas/patogenicidad , Encefalitis Transmitida por Garrapatas/virología , Fiebres Hemorrágicas Virales/virología , Macaca nemestrina , Animales , Chlorocebus aethiops , Citocinas/sangre , Virus de la Encefalitis Transmitidos por Garrapatas/genética , Virus de la Encefalitis Transmitidos por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/inmunología , Encefalitis Transmitida por Garrapatas/patología , Femenino , Células HEK293 , Fiebres Hemorrágicas Virales/inmunología , Fiebres Hemorrágicas Virales/patología , Humanos , Ganglios Linfáticos/virología , Células Vero , Viremia
10.
Front Immunol ; 12: 773146, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34956200

RESUMEN

Transcription factors (TFs) modulate genes involved in cell-type-specific proliferative and migratory properties, metabolic features, and effector functions. Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most important pathogen agents in the porcine industry; however, TFs have been poorly studied during the course of this disease. Therefore, we aimed to evaluate the expressions of the TFs T-bet, GATA3, FOXP3, and Eomesodermin (EOMES) in target organs (the lung, tracheobronchial lymph node, and thymus) and those of different effector cytokines (IFNG, TNFA, and IL10) and the Fas ligand (FASL) during the early phase of infection with PRRSV-1 strains of different virulence. Target organs from mock-, virulent Lena-, and low virulent 3249-infected animals humanely euthanized at 1, 3, 6, 8, and 13 days post-infection (dpi) were collected to analyze the PRRSV viral load, histopathological lesions, and relative quantification through reverse transcription quantitative PCR (RT-qPCR) of the TFs and cytokines. Animals belonging to both infected groups, but mainly those infected with the virulent Lena strain, showed upregulation of the TFs T-bet, EOMES, and FOXP3, together with an increase of the cytokine IFN-γ in target organs at the end of the study (approximately 2 weeks post-infection). These results are suggestive of a stronger polarization to Th1 cells and regulatory T cells (Tregs), but also CD4+ cytotoxic T lymphocytes (CTLs), effector CD8+ T cells, and γδT cells in virulent PRRSV-1-infected animals; however, their biological functionality should be the object of further studies.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Síndrome Respiratorio y de la Reproducción Porcina/inmunología , Proteínas de Dominio T Box/inmunología , Animales , Citocinas/genética , Citocinas/inmunología , Factor de Transcripción GATA3/inmunología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ganglios Linfáticos/virología , Síndrome Respiratorio y de la Reproducción Porcina/patología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Virus del Síndrome Respiratorio y Reproductivo Porcino , Porcinos , Proteínas de Dominio T Box/genética , Linfocitos T/inmunología , Timo/inmunología , Timo/patología , Timo/virología , Carga Viral , Virulencia
11.
Front Immunol ; 12: 757302, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790199

RESUMEN

Roles for viral infections and aberrant immune responses in driving localized neuroinflammation and neurodegeneration in multiple sclerosis (MS) are the focus of intense research. Epstein-Barr virus (EBV), as a persistent and frequently reactivating virus with major immunogenic influences and a near 100% epidemiological association with MS, is considered to play a leading role in MS pathogenesis, triggering localized inflammation near or within the central nervous system (CNS). This triggering may occur directly via viral products (RNA and protein) and/or indirectly via antigenic mimicry involving B-cells, T-cells and cytokine-activated astrocytes and microglia cells damaging the myelin sheath of neurons. The genetic MS-risk factor HLA-DR2b (DRB1*1501ß, DRA1*0101α) may contribute to aberrant EBV antigen-presentation and anti-EBV reactivity but also to mimicry-induced autoimmune responses characteristic of MS. A central role is proposed for inflammatory EBER1, EBV-miRNA and LMP1 containing exosomes secreted by viable reactivating EBV+ B-cells and repetitive release of EBNA1-DNA complexes from apoptotic EBV+ B-cells, forming reactive immune complexes with EBNA1-IgG and complement. This may be accompanied by cytokine- or EBV-induced expression of human endogenous retrovirus-W/-K (HERV-W/-K) elements and possibly by activation of human herpesvirus-6A (HHV-6A) in early-stage CNS lesions, each contributing to an inflammatory cascade causing the relapsing-remitting neuro-inflammatory and/or progressive features characteristic of MS. Elimination of EBV-carrying B-cells by antibody- and EBV-specific T-cell therapy may hold the promise of reducing EBV activity in the CNS, thereby limiting CNS inflammation, MS symptoms and possibly reversing disease. Other approaches targeting HHV-6 and HERV-W and limiting inflammatory kinase-signaling to treat MS are also being tested with promising results. This article presents an overview of the evidence that EBV, HHV-6, and HERV-W may have a pathogenic role in initiating and promoting MS and possible approaches to mitigate development of the disease.


Asunto(s)
Retrovirus Endógenos/patogenicidad , Herpesvirus Humano 4/patogenicidad , Herpesvirus Humano 6/patogenicidad , Esclerosis Múltiple/etiología , Enfermedades Neuroinflamatorias/virología , Anticuerpos Antivirales/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Autoinmunidad , Linfocitos B/inmunología , Barrera Hematoencefálica , Encéfalo/virología , Coinfección , ADN Viral/inmunología , Retrovirus Endógenos/fisiología , Antígenos Nucleares del Virus de Epstein-Barr/genética , Antígenos Nucleares del Virus de Epstein-Barr/inmunología , Productos del Gen env/fisiología , Predisposición Genética a la Enfermedad , Infecciones por Herpesviridae/complicaciones , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 4/inmunología , Herpesvirus Humano 6/inmunología , Humanos , Ganglios Linfáticos/virología , Modelos Inmunológicos , Imitación Molecular , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/terapia , Esclerosis Múltiple/virología , Vaina de Mielina/inmunología , Vaina de Mielina/patología , Enfermedades Neuroinflamatorias/etiología , Proteínas Gestacionales/fisiología , Activación Transcripcional , Activación Viral , Latencia del Virus
12.
Am J Trop Med Hyg ; 106(1): 182-186, 2021 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-34695799

RESUMEN

Rift Valley fever phlebovirus (RVFV) is a mosquito-transmitted phlebovirus (Family: Phenuiviridae, Order: Bunyavirales) causing severe neonatal mortality and abortion primarily in domestic ruminants. The susceptibility of young domestic swine to RVFV and this species' role in geographic expansion and establishment of viral endemicity is unclear. Six commercially bred Landrace-cross piglets were inoculated subcutaneously with 105 plaque-forming units of RVFV ZH501 strain and two piglets received a sham inoculum. All animals were monitored for clinical signs, viremia, viral shedding, and antibody response for 14 days. Piglets did not develop evidence of clinical disease, become febrile, or experience decreased weight gain during the study period. A brief lymphopenia followed by progressive lymphocytosis was observed following inoculation in all piglets. Four piglets developed a brief viremia for 2 days post-inoculation and three of these had detectable virus in oronasal secretions three days post-inoculation. Primary inoculated piglets seroconverted and those that developed detectable viremias had the highest titers assessed by serum neutralization (1:64-1:256). Two viremic piglets had a lymphoplasmacytic encephalitis with glial nodules; RVFV was not detected by immunohistochemistry in these sections. While young piglets do not appear to readily develop clinical disease following RVFV infection, results suggest swine could be subclinically infected with RVFV.


Asunto(s)
Fiebre del Valle del Rift/inmunología , Virus de la Fiebre del Valle del Rift/inmunología , Enfermedades de los Porcinos/virología , Animales , Encéfalo/patología , Encéfalo/virología , Susceptibilidad a Enfermedades , Femenino , Inmunohistoquímica , Hígado/patología , Hígado/virología , Ganglios Linfáticos/patología , Ganglios Linfáticos/virología , Masculino , ARN Viral/sangre , ARN Viral/genética , ARN Viral/aislamiento & purificación , Fiebre del Valle del Rift/sangre , Fiebre del Valle del Rift/transmisión , Fiebre del Valle del Rift/virología , Virus de la Fiebre del Valle del Rift/aislamiento & purificación , Virus de la Fiebre del Valle del Rift/patogenicidad , Bazo/patología , Bazo/virología , Sus scrofa , Porcinos , Enfermedades de los Porcinos/sangre , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/transmisión , Viremia/sangre , Viremia/inmunología , Viremia/virología
13.
Sci Rep ; 11(1): 19458, 2021 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-34593911

RESUMEN

Efficacious therapeutics for Ebola virus disease are in great demand. Ebola virus infections mediated by mucosal exposure, and aerosolization in particular, present a novel challenge due to nontypical massive early infection of respiratory lymphoid tissues. We performed a randomized and blinded study to compare outcomes from vehicle-treated and remdesivir-treated rhesus monkeys in a lethal model of infection resulting from aerosolized Ebola virus exposure. Remdesivir treatment initiated 4 days after exposure was associated with a significant survival benefit, significant reduction in serum viral titer, and improvements in clinical pathology biomarker levels and lung histology compared to vehicle treatment. These observations indicate that remdesivir may have value in countering aerosol-induced Ebola virus disease.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Adenosina Monofosfato/administración & dosificación , Adenosina Monofosfato/farmacología , Administración Intravenosa , Aerosoles , Alanina/administración & dosificación , Alanina/farmacología , Animales , Antivirales/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Fiebre Hemorrágica Ebola/sangre , Estimación de Kaplan-Meier , Hígado/efectos de los fármacos , Hígado/virología , Pulmón/patología , Pulmón/virología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Ganglios Linfáticos/virología , Macaca mulatta , Masculino , Distribución Aleatoria , Síndrome de Respuesta Inflamatoria Sistémica/tratamiento farmacológico , Síndrome de Respuesta Inflamatoria Sistémica/virología , Carga Viral/efectos de los fármacos , Viremia/tratamiento farmacológico
14.
EMBO J ; 40(22): e108966, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34618370

RESUMEN

Viremia in the vertebrate host is a major determinant of arboviral reservoir competency, transmission efficiency, and disease severity. However, immune mechanisms that control arboviral viremia are poorly defined. Here, we identify critical roles for the scavenger receptor MARCO in controlling viremia during arthritogenic alphavirus infections in mice. Following subcutaneous inoculation, arthritogenic alphavirus particles drain via the lymph and are rapidly captured by MARCO+ lymphatic endothelial cells (LECs) in the draining lymph node (dLN), limiting viral spread to the bloodstream. Upon reaching the bloodstream, alphavirus particles are cleared from the circulation by MARCO-expressing Kupffer cells in the liver, limiting viremia and further viral dissemination. MARCO-mediated accumulation of alphavirus particles in the draining lymph node and liver is an important host defense mechanism as viremia and viral tissue burdens are elevated in MARCO-/- mice and disease is more severe. In contrast to prior studies implicating a key role for lymph node macrophages in limiting viral dissemination, these findings exemplify a previously unrecognized arbovirus-scavenging role for lymphatic endothelial cells and improve our mechanistic understanding of viremia control during arthritogenic alphavirus infection.


Asunto(s)
Infecciones por Alphavirus/virología , Ganglios Linfáticos/citología , Receptores Inmunológicos/metabolismo , Viremia/patología , Alphavirus/patogenicidad , Animales , Fiebre Chikungunya/genética , Fiebre Chikungunya/virología , Células Endoteliales/virología , Interacciones Huésped-Patógeno , Macrófagos del Hígado/virología , Ganglios Linfáticos/virología , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , ARN Viral/metabolismo , Receptores Inmunológicos/genética , Análisis de la Célula Individual , Viremia/virología
15.
Arch Virol ; 166(11): 3127-3141, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34529151

RESUMEN

Porcine reproductive and respiratory syndrome (PRRS) is a highly contagious infectious disease caused by porcine reproductive and respiratory syndrome virus (PRRSV), which inflicts major economic losses on the global pig farming industry. Based on its similarity to highly pathogenic strains, the GDzj strain isolated in this study was predicted to be highly pathogenic. We therefore analyzed the pathogenicity of this strain experimentally in piglets. All piglets challenged with this virus experienced fever or high fever, loss of appetite, decreased food intake, daily weight loss, shortness of breath, and listlessness, and the necropsy results showed that they had experienced severe interstitial pneumonia. We then used the BAC system to construct a full-length cDNA infectious clone of GDzj, and the rescued virus displayed in vitro proliferation characteristics similar to those of the parental PRRSV strain. In summary, we successfully isolated a highly pathogenic PRRSV strain and constructed a full-length infectious cDNA clone from it, thereby providing an effective reverse genetics platform for further study of viral pathogenesis.


Asunto(s)
Síndrome Respiratorio y de la Reproducción Porcina/etiología , Virus del Síndrome Respiratorio y Reproductivo Porcino/genética , Virus del Síndrome Respiratorio y Reproductivo Porcino/patogenicidad , Animales , Cromosomas Artificiales Bacterianos , ADN Complementario/genética , Genoma Viral , Pulmón/virología , Ganglios Linfáticos/patología , Ganglios Linfáticos/virología , Filogenia , Neumonía Viral/patología , Neumonía Viral/virología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Virus del Síndrome Respiratorio y Reproductivo Porcino/crecimiento & desarrollo , Virus del Síndrome Respiratorio y Reproductivo Porcino/aislamiento & purificación , Porcinos
16.
Viruses ; 13(8)2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34452415

RESUMEN

The emergence and ensuing dominance of COVID-19 on the world stage has emphasized the urgency of efficient animal models for the development of therapeutics for and assessment of immune responses to SARS-CoV-2 infection. Shortcomings of current animal models for SARS-CoV-2 include limited lower respiratory disease, divergence from clinical COVID-19 disease, and requirements for host genetic modifications to permit infection. In this study, n = 12 specific-pathogen-free domestic cats were infected intratracheally with SARS-CoV-2 to evaluate clinical disease, histopathologic lesions, and viral infection kinetics at 4 and 8 days post-inoculation; n = 6 sham-inoculated cats served as controls. Intratracheal inoculation of SARS-CoV-2 produced a significant degree of clinical disease (lethargy, fever, dyspnea, and dry cough) consistent with that observed in the early exudative phase of COVID-19. Pulmonary lesions such as diffuse alveolar damage, hyaline membrane formation, fibrin deposition, and proteinaceous exudates were also observed with SARS-CoV-2 infection, replicating lesions identified in people hospitalized with ARDS from COVID-19. A significant correlation was observed between the degree of clinical disease identified in infected cats and pulmonary lesions. Viral loads and ACE2 expression were also quantified in nasal turbinates, distal trachea, lungs, and other organs. Results of this study validate a feline model for SARS-CoV-2 infection that results in clinical disease and histopathologic lesions consistent with acute COVID-19 in humans, thus encouraging its use for future translational studies.


Asunto(s)
COVID-19 , Gatos , Modelos Animales de Enfermedad , SARS-CoV-2/fisiología , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/patología , COVID-19/fisiopatología , COVID-19/virología , Femenino , Genoma Viral , Humanos , Pulmón/enzimología , Pulmón/patología , Pulmón/virología , Ganglios Linfáticos/virología , Masculino , ARN Viral/análisis , SARS-CoV-2/genética , Organismos Libres de Patógenos Específicos , Tráquea/enzimología , Tráquea/virología , Cornetes Nasales/enzimología , Cornetes Nasales/virología
17.
Viruses ; 13(7)2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34372502

RESUMEN

Squamous cell carcinoma of unknown primary (SCCUP) is a challenging diagnostic subgroup of oropharyngeal squamous cell carcinoma (OPSCC). The incidence of SCCUP is increasing in parallel with the well-documented increase in OPSCC and is likewise driven by the increase in human papillomavirus (HPV). The SCCUP patient often presents with a cystic lymph node metastasis and undergoes an aggressive diagnostic and treatment program. Detection of HPV in cytologic specimens indicates an oropharyngeal primary tumor origin and can guide the further diagnostic strategy. Advances in diagnostic modalities, e.g., transoral robotic surgery and transoral laser microsurgery, have increased the successful identification of the primary tumor site in HPV-induced SCCUP, and this harbors a potential for de-escalation treatment and increased survival. This review provides an overview of HPV-induced SCCUP, diagnostic modalities, and treatment options.


Asunto(s)
Alphapapillomavirus/patogenicidad , Carcinoma de Células Escamosas/virología , Neoplasias de Cabeza y Cuello/virología , Neoplasias Orofaríngeas/virología , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/patología , Humanos , Ganglios Linfáticos/virología , Metástasis Linfática , Neoplasias Orofaríngeas/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/virología
18.
Viruses ; 13(7)2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34372615

RESUMEN

Lymphopenia is a frequent hematological manifestation, associated with a severe course of COVID-19, with an insufficiently understood pathogenesis. We present molecular genetic immunohistochemical, and electron microscopic data on SARS-CoV-2 dissemination and viral load (VL) in lungs, mediastinum lymph nodes, and the spleen of 36 patients who died from COVID-19. Lymphopenia <1 × 109/L was observed in 23 of 36 (63.8%) patients. In 12 of 36 cases (33%) SARS-CoV-2 was found in lung tissues only with a median VL of 239 copies (range 18-1952) SARS-CoV-2 cDNA per 100 copies of ABL1. Histomorphological changes corresponding to bronchopneumonia and the proliferative phase of DAD were observed in these cases. SARS-CoV-2 dissemination into the lungs, lymph nodes, and spleen was detected in 23 of 36 patients (58.4%) and was associated with the exudative phase of DAD in most of these cases. The median VL in the lungs was 12,116 copies (range 810-250281), lymph nodes-832 copies (range 96-11586), and spleen-71.5 copies (range 0-2899). SARS-CoV-2 in all cases belonged to the 19A strain. A immunohistochemical study revealed SARS-CoV-2 proteins in pneumocytes, alveolar macrophages, and bronchiolar epithelial cells in lung tissue, sinus histiocytes of lymph nodes, as well as cells of the Billroth pulp cords and spleen capsule. SARS-CoV-2 particles were detected by transmission electron microscopy in the cytoplasm of the endothelial cell, macrophages, and lymphocytes. The infection of lymphocytes with SARS-CoV-2 that we discovered for the first time may indicate a possible link between lymphopenia and SARS-CoV-2-mediated cytotoxic effect.


Asunto(s)
COVID-19/virología , Pulmón/virología , Ganglios Linfáticos/virología , Linfopenia/virología , Mediastino/virología , SARS-CoV-2/aislamiento & purificación , Bazo/virología , Anciano , Anciano de 80 o más Años , Prueba de COVID-19 , Femenino , Humanos , Inmunohistoquímica , Pulmón/patología , Linfopenia/inmunología , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa Multiplex , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética , Carga Viral
19.
J Adv Res ; 31: 137-153, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34194838

RESUMEN

Introduction: Wild-type adult mice with intact interferon (IFN) system were neither susceptible to bluetongue virus (BTV) infection nor showed signs of morbidity/mortality. Establishment of immunologically competent wild-type adult mouse model with type I IFNs blockade is necessary to assess the pathogenesis, immune responses and testing of BTV vaccines. Objectives: Present study aimed to establish and characterize BTV serotype 1 infection in immunocompetent adult mice with type I IFNs blockade at the time of infection by studying immune responses and sequential pathology. Methods: Adult mice were administered with anti-mouse IFN-α/ß receptor subunit-1 (IFNAR1) blocking antibody (Clone: MAR1-5A3) 24 h before and after BTV serotype 1 infection, and sacrificed at various time points. Sequential pathology, BTV localization by immunohistochemistry and quantification by qRT-PCR, immune cell kinetics and apoptosis by flow cytometry, and cytokines estimation by c-ELISA and qRT-PCR were studied. Results: IFNAR blocked-infected mice developed clinical signs and typical lesions of BT; whereas, isotype-infected control mice did not develop any disease. The IFNAR blocked-infected mice showed enlarged, edematous, and congested lymph nodes (LNs) and spleen, and vascular (congestion and hemorrhage) and pneumonic lesions in lungs. Histopathologically, marked lymphoid depletion with "starry-sky pattern" due to lymphocytes apoptosis was noticed in the LNs and spleen. BTV antigen was detected and quantified in lymphoid organs, lungs, and other organs at various time points. Initial leukopenia (increased CD4+/CD8+ T cells ratio) followed by leukocytosis (decreased CD4+/CD8+ T cells ratio) and significantly increased biochemical values were noticed in IFNAR blocked-infected mice. Increased apoptotic cells in PBMCs and tissues coincided with viral load and levels of different cytokines in blood, spleen and draining LNs and notably varied between time points in IFNAR blocked-infected mice. Conclusion: Present study is first to characterize BTV serotype 1 infection in immunocompetent adult mouse with type I IFNs blockade. The findings will be useful for studying pathogenesis and testing the efficacy of BTV vaccines.


Asunto(s)
Virus de la Lengua Azul/genética , Lengua Azul/inmunología , Lengua Azul/patología , Interferón Tipo I/inmunología , Animales , Anticuerpos Bloqueadores/inmunología , Apoptosis , Virus de la Lengua Azul/inmunología , Femenino , Leucocitos/inmunología , Leucocitosis/inmunología , Leucopenia/inmunología , Pulmón/patología , Pulmón/virología , Ganglios Linfáticos/patología , Ganglios Linfáticos/virología , Ratones , Modelos Inmunológicos , Receptor de Interferón alfa y beta/inmunología , Serogrupo , Ovinos , Bazo/patología , Bazo/virología , Vacunas Virales/inmunología
20.
PLoS One ; 16(7): e0254872, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34280238

RESUMEN

BACKGROUND: COVID-19 is only partly understood, and the level of evidence available in terms of pathophysiology, epidemiology, therapy, and long-term outcome remains limited. During the early phase of the pandemic, it was necessary to effectively investigate all aspects of this new disease. Autopsy can be a valuable procedure to investigate the internal organs with special techniques to obtain information on the disease, especially the distribution and type of organ involvement. METHODS: During the first wave of COVID-19 in Germany, autopsies of 19 deceased patients were performed. Besides gross examination, the organs were analyzed with standard histology and polymerase-chain-reaction for SARS-CoV-2. Polymerase chain reaction positive localizations were further analyzed with immunohistochemistry and RNA-in situ hybridization for SARS-CoV-2. RESULTS: Eighteen of 19 patients were found to have died due to COVID-19. Clinically relevant histological changes were only observed in the lungs. Diffuse alveolar damage in considerably different degrees was noted in 18 cases. Other organs, including the central nervous system, did not show specific micromorphological alterations. In terms of SARS-CoV-2 detection, the focus remains on the upper airways and lungs. This is true for both the number of positive samples and the viral load. A highly significant inverse correlation between the stage of diffuse alveolar damage and viral load was found on a case and a sample basis. Mediastinal lymph nodes and fat were also affected by the virus at high frequencies. By contrast, other organs rarely exhibited a viral infection. Moderate to strong correlations between the methods for detecting SARS-CoV-2 were observed for the lungs and for other organs. CONCLUSIONS: The lung is the most affected organ in gross examination, histology and polymerase chain reaction. SARS-CoV-2 detection in other organs did not reveal relevant or specific histological changes. Moreover, we did not find CNS involvement.


Asunto(s)
COVID-19/virología , Sistema Nervioso Central/virología , Pulmón/virología , Ganglios Linfáticos/virología , Carga Viral , Anciano , Anciano de 80 o más Años , Autopsia/estadística & datos numéricos , COVID-19/epidemiología , COVID-19/patología , Sistema Nervioso Central/patología , Femenino , Humanos , Pulmón/patología , Ganglios Linfáticos/patología , Masculino , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...