Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurovirol ; 30(1): 86-99, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38453879

RESUMEN

Simian varicella virus (SVV) produces peripheral inflammatory responses during varicella (primary infection) and zoster (reactivation) in rhesus macaques (RM). However, it is unclear if peripheral measures are accurate proxies for central nervous system (CNS) responses. Thus, we analyzed cytokine and Aß42/Aß40 changes in paired serum and cerebrospinal fluid (CSF) during the course of infection. During varicella and zoster, every RM had variable changes in serum and CSF cytokine and Aß42/Aß40 levels compared to pre-inoculation levels. Overall, peripheral infection appears to affect CNS cytokine and Aß42/Aß40 levels independent of serum responses, suggesting that peripheral disease may contribute to CNS disease.


Asunto(s)
Péptidos beta-Amiloides , Citocinas , Macaca mulatta , Animales , Péptidos beta-Amiloides/líquido cefalorraquídeo , Péptidos beta-Amiloides/sangre , Citocinas/líquido cefalorraquídeo , Citocinas/sangre , Activación Viral , Fragmentos de Péptidos/líquido cefalorraquídeo , Fragmentos de Péptidos/sangre , Varicellovirus/genética , Varicellovirus/inmunología , Herpesvirus Humano 3/patogenicidad , Herpesvirus Humano 3/inmunología , Infecciones por Herpesviridae/líquido cefalorraquídeo , Infecciones por Herpesviridae/virología , Infecciones por Herpesviridae/sangre , Infecciones por Herpesviridae/inmunología , Masculino , Herpes Zóster/líquido cefalorraquídeo , Herpes Zóster/virología , Herpes Zóster/sangre , Herpes Zóster/inmunología , Enfermedades de los Monos/virología , Enfermedades de los Monos/líquido cefalorraquídeo , Enfermedades de los Monos/sangre
2.
J Infect Dis ; 2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-38135285

RESUMEN

Respiratory syncytial virus (RSV) primarily infects the respiratory epithelium, but growing evidence suggests it may also be responsible for neurological sequelae. In 3D microphysiological peripheral nerve cultures, RSV infected neurons, macrophages, and dendritic cells along two distinct trajectories depending on the initial viral load. Low-level infection was transient, primarily involved macrophages, and induced moderate chemokine release with transient neural hypersensitivity. Infection with higher viral loads was persistent, infected neuronal cells in addition to monocytes, and induced robust chemokine release followed by progressive neurotoxicity. In spinal cord cultures, RSV infected microglia and dendritic cells but not neurons, producing a moderate chemokine expression pattern. The persistence of infection was variable but could be identified in dendritic cells as long as 30 days post-inoculation. This study suggests that RSV can disrupt neuronal function directly through infection of peripheral neurons and indirectly through infection of resident monocytes, and inflammatory chemokines likely mediate both mechanisms.

3.
J Med Primatol ; 51(1): 20-26, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34778968

RESUMEN

BACKGROUND: Simian varicella virus (SVV) is a primate herpesvirus that causes a natural varicella-like disease in Old World monkeys and may cause epizootics in facilities housing nonhuman primates. SVV infection of nonhuman primates is used as an experimental model to investigate varicella pathogenesis and to develop antiviral strategies. METHODS: An indirect enzyme-linked immunosorbent assay (ELISA) was developed to detect SVV antibodies in infected rhesus macaque monkeys. RESULTS: An ELISA determined SVV antibody titers following experimental infection. SVV IgG was detected by day 14 post-infection and remained elevated for at least 84 days. CONCLUSIONS: The SVV ELISA is a safe and rapid approach to confirm SVV seropositivity and to determine SVV antibody titers in naturally and experimentally SVV-infected monkeys. In addition to being a useful diagnostic assay to rapidly confirm acute disease or past SVV infection, the SVV ELISA is a valuable epidemiological tool to determine the incidence of SVV in non-human primate facilities.


Asunto(s)
Varicela , Varicellovirus , Animales , Ensayo de Inmunoadsorción Enzimática , Herpesvirus Humano 3 , Macaca mulatta
4.
J Virol ; 93(3)2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30404798

RESUMEN

Rhesus macaques intrabronchially inoculated with simian varicella virus (SVV), the counterpart of human varicella-zoster virus (VZV), developed primary infection with viremia and rash, which resolved upon clearance of viremia, followed by the establishment of latency. To assess the role of CD4 T cell immunity in reactivation, monkeys were treated with a single 50-mg/kg dose of a humanized monoclonal anti-CD4 antibody; within 1 week, circulating CD4 T cells were reduced from 40 to 60% to 5 to 30% of the total T cell population and remained low for 2 months. Very low viremia was seen only in some of the treated monkeys. Zoster rash developed after 7 days in the monkey with the most extensive CD4 T cell depletion (5%) and in all other monkeys at 10 to 49 days posttreatment, with recurrent zoster in one treated monkey. SVV DNA was detected in the lung from two of five monkeys, in bronchial lymph nodes from one of the five monkeys, and in ganglia from at least two dermatomes in three of five monkeys. Immunofluorescence analysis of skin rash, lungs, lymph nodes, and ganglia revealed SVV ORF63 protein at the following sites: sweat glands in skin; type II cells in lung alveoli, macrophages, and dendritic cells in lymph nodes; and the neuronal cytoplasm of ganglia. Detection of SVV antigen in multiple tissues upon CD4 T cell depletion and virus reactivation suggests a critical role for CD4 T cell immunity in controlling varicella virus latency.IMPORTANCE Reactivation of latent VZV in humans can result in serious neurological complications. VZV-specific cell-mediated immunity is critical for the maintenance of latency. Similar to VZV in humans, SVV causes varicella in monkeys, establishes latency in ganglia, and reactivates to produce shingles. Here, we show that depletion of CD4 T cells in rhesus macaques results in SVV reactivation, with virus antigens found in zoster rash and SVV DNA and antigens found in lungs, lymph nodes, and ganglia. These results suggest the critical role of CD4 T cell immunity in controlling varicella virus latency.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Herpesviridae/inmunología , Depleción Linfocítica , Piel/inmunología , Varicellovirus/aislamiento & purificación , Activación Viral/inmunología , Latencia del Virus/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/virología , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/virología , Modelos Animales de Enfermedad , Femenino , Ganglios/citología , Ganglios/inmunología , Ganglios/virología , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Pulmón/citología , Pulmón/inmunología , Pulmón/virología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/virología , Macaca mulatta , Masculino , Piel/citología , Piel/virología
5.
J Neurovirol ; 26(6): 945-951, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32964407

RESUMEN

Varicella and zoster, produced by varicella-zoster virus (VZV), are associated with an increased risk of stroke that may be due to persistent inflammation and hypercoagulability. Because substance P is associated with inflammation, hypercoagulability, and atherosclerotic plaque rupture that may contribute to increased stroke risk after VZV infection, we measured serum substance P in simian varicella virus-infected rhesus macaques. We found significantly increased and persistent serum substance P concentrations during varicella and zoster compared with pre-inoculation, supporting the hypothesis that VZV-induced increases in serum substance P may contribute to increased stroke risk associated with VZV infection.


Asunto(s)
Herpesvirus Humano 3/inmunología , Sustancia P/genética , Infección por el Virus de la Varicela-Zóster/inmunología , Infección por el Virus de la Varicela-Zóster/veterinaria , Activación Viral/inmunología , Animales , Biomarcadores/sangre , Expresión Génica , Herpesvirus Humano 3/patogenicidad , Inmunosupresores/administración & dosificación , Inflamación , Macaca mulatta , Masculino , Riesgo , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/veterinaria , Sustancia P/sangre , Sustancia P/inmunología , Tacrolimus/administración & dosificación , Infección por el Virus de la Varicela-Zóster/complicaciones , Infección por el Virus de la Varicela-Zóster/genética , Irradiación Corporal Total
6.
J Gen Virol ; 100(1): 26-34, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30480508

RESUMEN

For an effective T-cell activation and response, co-stimulation is required in addition to the antigen-specific signal from their antigen receptors. The CD2/CD58 interaction is considered as one of the most important T-cell co-stimulatory pathways for T-cell activation and proliferation, and its role in regulating intestinal T-cell function in acute and chronic SIV -infected macaques is poorly documented. Here, we demonstrated a significant reduction of CD58 expression in both T- and B-cell populations during acute SIV infection along with high plasma viral load and a loss of intestinal CD4+ T cells compared to SIV-uninfected control macaques. The reduction of CD58 expression in T cells was correlated with the reduced expression of T-cell-mediated IL-2 and TNFα production. Together, these results indicate that reduction in the CD2/CD58 interaction pathway in mucosal lymphocytes might play a crucial role in mucosal T-cell dysfunction during acute SIV/HIV infection.


Asunto(s)
Antígenos CD58/biosíntesis , Expresión Génica , Interleucina-2/metabolismo , Mucosa Intestinal/patología , Linfocitos Intraepiteliales/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Linfocitos B/inmunología , Activación de Linfocitos , Macaca , Plasma/virología , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación , Carga Viral
7.
J Virol ; 92(7)2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29343566

RESUMEN

Simian varicella virus (SVV), the primate counterpart of varicella-zoster virus, causes varicella (chickenpox), establishes latency in ganglia, and reactivates to produce zoster. We previously demonstrated that a recombinant SVV expressing enhanced green fluorescent protein (rSVV.eGFP) is slightly attenuated both in culture and in infected monkeys. Here, we generated two additional recombinant SVVs to visualize infected cells in vitro and in vivo One harbors eGFP fused to the N terminus of open reading frame 9 (ORF9) (rSVV.eGFP-2a-ORF9), and another harbors eGFP fused to the C terminus of ORF66 (rSVV.eGFP-ORF66). Both recombinant viruses efficiently expressed eGFP in cultured cells. Both recombinant SVV infections in culture were comparable to that of wild-type SVV (SVV.wt). Unlike SVV.wt, eGFP-tagged SVV did not replicate in rhesus cells in culture. Intratracheal (i.t.) or i.t. plus intravenous (i.v.) inoculation of rhesus macaques with these new eGFP-tagged viruses resulted in low viremia without varicella rash, although SVV DNA was abundant in bronchoalveolar lavage (BAL) fluid at 10 days postinoculation (dpi). SVV DNA was also found in trigeminal ganglia of one monkey inoculated with rSVV.eGFP-ORF66. Intriguingly, a humoral response to both SVV and eGFP was observed. In addition, monkeys inoculated with the eGFP-expressing viruses were protected from superinfection with SVV.wt, suggesting that the monkeys had mounted an efficient immune response. Together, our results show that eGFP expression could be responsible for their reduced pathogenesis.IMPORTANCE SVV infection in nonhuman primates has served as an extremely useful animal model to study varicella-zoster virus (VZV) pathogenesis. eGFP-tagged viruses are a great tool to investigate their pathogenesis. We constructed and tested two new recombinant SVVs with eGFP inserted into two different locations in the SVV genome. Both recombinant SVVs showed robust replication in culture but reduced viremia compared to that with SVV.wt during primary infection in rhesus macaques. Our results indicate that conclusions on eGFP-tagged viruses based on in vitro results should be handled with care, since eGFP expression could result in attenuation of the virus.


Asunto(s)
Regulación Viral de la Expresión Génica , Proteínas Fluorescentes Verdes , Infecciones por Herpesviridae , Enfermedades de los Monos , Sistemas de Lectura Abierta , Varicellovirus , Animales , Línea Celular , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/veterinaria , Macaca mulatta , Enfermedades de los Monos/genética , Enfermedades de los Monos/metabolismo , Enfermedades de los Monos/patología , Varicellovirus/genética , Varicellovirus/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
8.
J Virol ; 89(19): 9817-24, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26178993

RESUMEN

UNLABELLED: Like varicella-zoster virus (VZV), simian varicella virus (SVV) reactivates to produce zoster. In the present study, 5 rhesus macaques were inoculated intrabronchially with SVV, and 5 months later, 4 monkeys were immunosuppressed; 1 monkey was not immunosuppressed but was subjected to the stress of transportation. In 4 monkeys, a zoster rash developed 7 to 12 weeks after immunosuppression, and a rash also developed in the monkey that was not immunosuppressed. Analysis at 24 to 48 h after zoster revealed SVV antigen in the lung alveolar wall, in ganglionic neurons and nonneuronal cells, and in skin and in lymph nodes. In skin, SVV was found primarily in sweat glands. In lymph nodes, the SVV antigen colocalized mostly with macrophages, dendritic cells, and, to a lesser extent, T cells. The presence of SVV in lymph nodes, as verified by quantitative PCR detection of SVV DNA, might reflect the sequestration of virus by macrophages and dendritic cells in lymph nodes or the presentation of viral antigens to T cells to initiate an immune response against SVV, or both. IMPORTANCE: VZV causes varicella (chickenpox), becomes latent in ganglia, and reactivates to produce zoster and multiple other serious neurological disorders. SVV in nonhuman primates has proved to be a useful model in which the pathogenesis of the virus parallels the pathogenesis of VZV in humans. Here, we show that SVV antigens are present in sweat glands in skin and in macrophages and dendritic cells in lymph nodes after SVV reactivation in monkeys, raising the possibility that macrophages and dendritic cells in lymph nodes serve as antigen-presenting cells to activate T cell responses against SVV after reactivation.


Asunto(s)
Herpes Zóster/patología , Herpes Zóster/virología , Ganglios Linfáticos/virología , Varicellovirus/fisiología , Activación Viral/fisiología , Animales , Chlorocebus aethiops , ADN Viral/análisis , Células Dendríticas/virología , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Terapia de Inmunosupresión , Ganglios Linfáticos/citología , Macaca mulatta , Macrófagos/virología , Reacción en Cadena en Tiempo Real de la Polimerasa , Piel/patología , Piel/virología , Linfocitos T/virología , Células Vero
9.
J Virol ; 87(5): 2979-82, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23269790

RESUMEN

Ganglia of monkeys with reactivated simian varicella virus (SVV) contained more CD8 than CD4 T cells around neurons. The abundance of CD8 T cells was greater less than 2 months after reactivation than that at later times and correlated with that of CXCL10 RNA but not with those of SVV protein or open reading frame 61 (ORF61) antisense RNA. CXCL10 RNA colocalized with T-cell clusters. After SVV reactivation, transient T-cell infiltration, possibly mediated by CXCL10, parallels varicella zoster virus (VZV) reactivation in humans.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Quimiocina CXCL10/metabolismo , Ganglios/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Neuronas/inmunología , Varicellovirus/inmunología , Varicellovirus/metabolismo , Varicellovirus/fisiología , Animales , Linfocitos T CD8-positivos/metabolismo , ADN Viral/genética , Ganglios/metabolismo , Ganglios/virología , Infecciones por Herpesviridae/genética , Macaca fascicularis , Neuronas/virología , Sistemas de Lectura Abierta , ARN sin Sentido/biosíntesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Varicellovirus/genética , Carga Viral , Activación Viral , Latencia del Virus/genética
10.
J Neurovirol ; 20(3): 309-13, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24549971

RESUMEN

Like varicella zoster virus in humans, simian varicella virus (SVV) becomes latent in ganglionic neurons along the entire neuraxis and reactivates in immunosuppressed monkeys. Five rhesus macaques were inoculated with SVV; 142 days later (latency), four monkeys were immunosuppressed, and T cells were analyzed for naïve, memory, and effector phenotypes and expression of programmed death receptor-1 (PD-1; T cell exhaustion). All T cell subsets decreased during immunosuppression and except for CD8 effectors, peaked 2 weeks before zoster. Compared to before immunosuppression, PD-1 expression increased at reactivation. Increased T cells before zoster is likely due to virus reactivation.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/virología , Herpes Zóster/inmunología , Herpesvirus Humano 3/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Envejecimiento/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Glucocorticoides/farmacología , Herpes Zóster/patología , Memoria Inmunológica/inmunología , Inmunosupresores/farmacología , Macaca mulatta , Prednisona/farmacología , Activación Viral/inmunología
11.
J Neurovirol ; 20(5): 526-30, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25139181

RESUMEN

Simian varicella virus (SVV) infection of non-human primates models human varicella zoster virus (VZV) infection. Assessment of cell signaling immune responses in monkeys after primary SVV infection, after immunosuppression and during reactivation revealed strong pro-inflammatory responses and lesser anti-inflammatory components during varicella and reactivation. Pro-inflammatory mediators elevated during varicella included interferon-gamma (IFN-γ), interleukin (IL)-6, monocyte chemoattractant protein (MCP-1), interferon inducible T-cell α chemoattractant protein (I-TAC), interferon processing protein (IP-10), and anti-inflammatory interleukin-1 Receptor antagonist (IL-1Ra). After immunosuppression and at reactivation, levels of pro-inflammatory mediators MCP-1, eotaxin, IL-6, IL-8, MIF, RANTES (regulated-on-activation normal T-cell expressed and secreted), and HGF (hepatocyte growth factor) were elevated, as was the anti-inflammatory mediator IL-1Ra. Characterization of cytokine, chemokine and growth factor responses during different stages of varicella virus infection will facilitate immunotherapeutic and vaccine strategies.


Asunto(s)
Infecciones por Herpesviridae/inmunología , Activación Viral/inmunología , Latencia del Virus/inmunología , Animales , Quimiocinas/inmunología , Modelos Animales de Enfermedad , Inflamación/inmunología , Inflamación/virología , Macaca mulatta , Masculino , Varicellovirus/fisiología
12.
Res Sq ; 2023 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-37886544

RESUMEN

Simian varicella virus (SVV) produces peripheral inflammatory responses during varicella (primary infection) and zoster (reactivation) in rhesus macaques (RM). However, it is unclear if peripheral measures are accurate proxies for central nervous system (CNS) responses. Thus, we analyzed cytokine and Aß42/Aß40 changes in paired serum and cerebrospinal fluid (CSF) during the course of infection. During varicella and zoster, every RM had variable changes in serum and CSF cytokine and Aß42/Aß40 levels compared to pre-inoculation levels. Overall, peripheral infection appears to affect CNS cytokine and Aß42/Aß40 levels independent of serum responses, suggesting that peripheral disease may contribute to CNS disease.

13.
Commun Biol ; 6(1): 1265, 2023 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-38092883

RESUMEN

SARS-CoV-2 infection can cause persistent respiratory sequelae. However, the underlying mechanisms remain unclear. Here we report that sub-lethally infected K18-human ACE2 mice show patchy pneumonia associated with histiocytic inflammation and collagen deposition at 21 and 45 days post infection (DPI). Transcriptomic analyses revealed that compared to influenza-infected mice, SARS-CoV-2-infected mice had reduced interferon-gamma/alpha responses at 4 DPI and failed to induce keratin 5 (Krt5) at 6 DPI in lung, a marker of nascent pulmonary progenitor cells. Histologically, influenza- but not SARS-CoV-2-infected mice showed extensive Krt5+ "pods" structure co-stained with stem cell markers Trp63/NGFR proliferated in the pulmonary consolidation area at both 7 and 14 DPI, with regression at 21 DPI. These Krt5+ "pods" structures were not observed in the lungs of SARS-CoV-2-infected humans or nonhuman primates. These results suggest that SARS-CoV-2 infection fails to induce nascent Krt5+ cell proliferation in consolidated regions, leading to incomplete repair of the injured lung.


Asunto(s)
COVID-19 , Gripe Humana , Ratones , Humanos , Animales , SARS-CoV-2 , Pulmón , Perfilación de la Expresión Génica
14.
J Neurovirol ; 18(2): 91-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22399159

RESUMEN

Varicella-zoster virus (VZV) causes varicella (chickenpox), becomes latent in ganglia along the entire neuraxis, and may reactivate to cause herpes zoster (shingles). VZV may infect ganglia via retrograde axonal transport from infected skin or through hematogenous spread. Simian varicella virus (SVV) infection of rhesus macaques provides a useful model system to study the pathogenesis of human VZV infection. To dissect the virus and host immune factors during acute SVV infection, we analyzed four SVV-seronegative Chinese rhesus macaques infected intratracheally with cell-associated 5 × 10³ plaque-forming units (pfu) of SVV-expressing green fluorescent protein (n = 2) or 5 × 104 pfu of wild-type SVV (n = 2). All monkeys developed viremia and SVV-specific adaptive B- and T-cell immune responses, but none developed skin rash. At necropsy 21 days postinfection, SVV DNA was found in ganglia along the entire neuraxis and in viscera, and SVV RNA was found in ganglia, but not in viscera. The amount of SVV inoculum was associated with the extent of viremia and the immune response to virus. Our findings demonstrate that acute SVV infection of Chinese rhesus macaques leads to ganglionic infection by the hematogenous route and the induction of a virus-specific adaptive memory response in the absence of skin rash.


Asunto(s)
Varicela/virología , Ganglios/virología , Herpesvirus Humano 3/fisiología , Viremia/virología , Inmunidad Adaptativa , Animales , Linfocitos B/inmunología , Linfocitos B/virología , Varicela/inmunología , Varicela/patología , ADN Viral/biosíntesis , Modelos Animales de Enfermedad , Exantema , Ganglios/inmunología , Ganglios/patología , Humanos , Macaca mulatta , Piel/virología , Linfocitos T/inmunología , Linfocitos T/virología , Viremia/inmunología , Viremia/patología , Latencia del Virus
15.
Virol J ; 9: 160, 2012 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-22889373

RESUMEN

BACKGROUND: An effective AIDS vaccine remains one of the highest priorities in HIV-research. Our recent study showed that vaccination of rhesus macaques with recombinant simian varicella virus (rSVV) vector - simian immunodeficiency virus (SIV) envelope and gag genes, induced neutralizing antibodies and cellular immune responses to SIV and also significantly reduced plasma viral loads following intravenous pathogenic challenge with SIVMAC251/CX1. FINDINGS: The purpose of this study was to define cellular immunological correlates of protection in rSVV-SIV vaccinated and SIV challenged animals. Immunofluorescent staining and multifunctional assessment of SIV-specific T-cell responses were evaluated in both Experimental and Control vaccinated animal groups. Significant increases in the proliferating CD4+ T-cell population and polyfunctional T-cell responses were observed in all Experimental-vaccinated animals compared with the Control-vaccinated animals. CONCLUSIONS: Increased CD4+ T-cell proliferation was significantly and inversely correlated with plasma viral load. Increased SIV-specific polyfunctional cytokine responses and increased proliferation of CD4+ T-cell may be crucial to control plasma viral loads in vaccinated and SIVMAC251/CX1 challenged macaques.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Vacunas contra el SIDAS/inmunología , Virus de la Inmunodeficiencia de los Simios/genética , Varicellovirus/genética , Carga Viral , Viremia/prevención & control , Animales , Proliferación Celular , Citocinas/metabolismo , Macaca mulatta , Plasma/virología , Vacunas contra el SIDAS/administración & dosificación , Vacunas contra el SIDAS/genética , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
16.
Viruses ; 14(6)2022 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-35746639

RESUMEN

Primary simian varicella virus (SVV) infection and reactivation in nonhuman primates is a valuable animal model in the study of varicella zoster virus disease [varicella (chickenpox) and herpes zoster (shingles)]. To understand SVV pathogenesis in skin, we inoculated 10 rhesus macaques with SVV, resulting in varicella rash. After the establishment of latency, eight of the monkeys were immunosuppressed using tacrolimus with or without irradiation and prednisone and two monkeys were not immunosuppressed. Zoster rash developed in all immunosuppressed monkeys and in one non-immunosuppressed monkey. Five monkeys had recurrent zoster. During varicella and zoster, SVV DNA in skin scrapings ranged from 50 to 107 copies/100 ng of total DNA and 2-127 copies/100 ng of total DNA, respectively. Detection of SVV DNA in blood during varicella was more frequent and abundant compared to that of zoster. During varicella and zoster, SVV antigens colocalized with neurons expressing ß-III tubulin in epidermis, hair follicles, and sweat glands, suggesting axonal transport of the virus. Together, we have demonstrated that both SVV DNA and antigens can be detected in skin lesions during varicella and zoster, providing the basis for further studies on SVV skin pathogenesis, including immune responses and mechanisms of peripheral spread.


Asunto(s)
Varicela , Exantema , Herpes Zóster , Varicellovirus , Animales , Herpesvirus Humano 3/fisiología , Macaca mulatta , Varicellovirus/genética
17.
Viruses ; 14(12)2022 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-36560823

RESUMEN

HIV vaccine mediated efficacy, using an expanded live attenuated recombinant varicella virus-vectored SIV rSVV-SIVgag/env vaccine prime with adjuvanted SIV-Env and SIV-Gag protein boosts, was evaluated in a female rhesus macaques (RM) model against repeated intravaginal SIV challenges. Vaccination induced anti-SIV IgG responses and neutralizing antibodies were found in all vaccinated RMs. Three of the eight vaccinated RM remained uninfected (vaccinated and protected, VP) after 13 repeated challenges with the pathogenic SIVmac251-CX-1. The remaining five vaccinated and infected (VI) macaques had significantly reduced plasma viral loads compared with the infected controls (IC). A significant increase in systemic central memory CD4+ T cells and mucosal CD8+ effector memory T-cell responses was detected in vaccinated RMs compared to controls. Variability in lymph node SIV-Gag and Env specific CD4+ and CD8+ T cell cytokine responses were detected in the VI RMs while all three VP RMs had more durable cytokine responses following vaccination and prior to challenge. VI RMs demonstrated predominately SIV-specific monofunctional cytokine responses while the VP RMs generated polyfunctional cytokine responses. This study demonstrates that varicella virus-vectored SIV vaccination with protein boosts induces a 37.5% efficacy rate against pathogenic SIV challenge by generating mucosal memory, virus specific neutralizing antibodies, binding antibodies, and polyfunctional T-cell responses.


Asunto(s)
Varicela , Vacunas contra el SIDAS , Síndrome de Inmunodeficiencia Adquirida del Simio , Virus de la Inmunodeficiencia de los Simios , Animales , Femenino , Virus de la Inmunodeficiencia de los Simios/genética , Macaca mulatta , Vacunas Sintéticas/genética , Vacunas contra el SIDAS/genética , Anticuerpos Neutralizantes , Citocinas , Anticuerpos Antivirales
18.
J Virol ; 84(23): 12454-7, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20861271

RESUMEN

Studies of varicella-zoster virus gene expression during latency require the acquisition of human ganglia at autopsy. Concerns have been raised that the virus might reactivate immediately after death. Because features of varicella-zoster virus latency are similar in primate and human ganglia, we examined virus gene expression in tissues either processed immediately or kept at 4°C for 30 h before necropsy of two monkeys inoculated with simian varicella-zoster virus and euthanized 117 days later. Virus transcription and the detection of open reading frame (ORF) 63 protein in the cytoplasm of neurons were comparable. Thus, a 30-h delay after death did not affect varicella-zoster virus expression in latently infected ganglia.


Asunto(s)
Varicela/fisiopatología , Ganglios/metabolismo , Regulación Viral de la Expresión Génica/fisiología , Herpesvirus Humano 3/fisiología , Proteínas Inmediatas-Precoces/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Latencia del Virus/fisiología , Animales , Varicela/metabolismo , Herpesvirus Humano 3/metabolismo , Inmunohistoquímica , Macaca mulatta , Neuronas/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Viremia/sangre
19.
Viruses ; 13(7)2021 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-34206909

RESUMEN

Latent varicella zoster virus (VZV) has been detected in human adrenal glands, raising the possibility of virus-induced adrenal damage and dysfunction during primary infection or reactivation. Rare cases of bilateral adrenal hemorrhage and insufficiency associated with VZV reactivation have been reported. Since there is no animal model for VZV infection of adrenal glands, we obtained adrenal glands from two non-human primates (NHPs) that spontaneously developed varicella from primary simian varicella virus (SVV) infection, the NHP VZV homolog. Histological and immunohistochemical analysis revealed SVV antigen and DNA in the adrenal medulla and cortex of both animals. Adrenal glands were observed to have Cowdry A inclusion bodies, cellular necrosis, multiple areas of hemorrhage, and varying amounts of polymorphonuclear cells. No specific association of SVV antigen with ßIII-tubulin-positive nerve fibers was found. Overall, we found that SVV can productively infect NHP adrenal glands, and is associated with inflammation, hemorrhage, and cell death. These findings suggest that further studies are warranted to examine the contribution of VZV infection to human adrenal disease. This study also suggests that VZV infection may present itself as acute adrenal dysfunction with "long-hauler" symptoms of fatigue, weakness, myalgias/arthralgias, and hypotension.


Asunto(s)
Glándulas Suprarrenales/patología , Glándulas Suprarrenales/virología , Infecciones por Herpesviridae/patología , Herpesvirus Humano 3/patogenicidad , Glándulas Suprarrenales/citología , Animales , Femenino , Infecciones por Herpesviridae/virología , Técnicas Histológicas , Macaca fascicularis/virología , Masculino
20.
J Neurovirol ; 16(5): 342-54, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20822371

RESUMEN

Simian varicella virus (SVV) infection of primates resembles human varicella-zoster virus (VZV) infection. After primary infection, SVV becomes latent in ganglia and reactivates after immunosuppression or social and environmental stress. Herein, natural SVV infection was established in 5 cynomolgus macaques (cynos) and 10 African green (AG) monkeys. Four cynos were treated with the immunosuppressant tacrolimus (80 to 300 μg/kg/day) for 4 months and 1 was untreated (group 1). Four AG monkeys were exposed to a single dose (200 cGy) of x-irradiation (group 2), and 4 other AG monkeys were irradiated and treated with tacrolimus for 4 months (group 3); the remaining 2 AG monkeys were untreated. Zoster rash developed 1 to 2 weeks after tacrolimus treatment in 3 of 4 monkeys in group 1, 6 weeks after irradiation in 1 of 4 monkeys in group 2, and 1 to 2 weeks after irradiation in all 4 monkeys in group 3. All monkeys were euthanized 1 to 4 months after immunosuppression. SVV antigens were detected immunohistochemically in skin biopsies as well as in lungs of most monkeys. Low copy number SVV DNA was detected in ganglia from all three groups of monkeys, including controls. RNA specific for SVV ORFs 61, 63, and 9 was detected in ganglia from one immunosuppressed monkey in group 1. SVV antigens were detected in multiple ganglia from all immunosuppressed monkeys in every group, but not in controls. These results indicate that tacrolimus treatment produced reactivation in more monkeys than irradiation and tacrolimus and irradiation increased the frequency of SVV reactivation as compared to either treatment alone.


Asunto(s)
Varicela/inducido químicamente , Herpes Zóster/virología , Herpesvirus Humano 3/fisiología , Inmunosupresores/farmacología , Tacrolimus/farmacología , Activación Viral/efectos de los fármacos , Animales , Chlorocebus aethiops , Macaca fascicularis , Activación Viral/efectos de la radiación , Latencia del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA