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 Virol ; 98(7): e0021324, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38832789

RESUMEN

Human cytomegalovirus (HCMV) is a ß-herpesvirus that poses severe disease risk for immunocompromised patients who experience primary infection or reactivation. Development and optimization of safe and effective anti-HCMV therapeutics is of urgent necessity for the prevention and treatment of HCMV-associated diseases in diverse populations. The use of neutralizing monoclonal antibodies (mAbs) to limit HCMV infection poses a promising therapeutic strategy, as anti-HCMV mAbs largely inhibit infection by targeting virion glycoprotein complexes. In contrast, the small-molecule compounds currently approved for patients (e.g., ganciclovir, letermovir, and maribavir) target later stages of the HCMV life cycle. Here, we present a broadly neutralizing human mAb, designated 1C10, elicited from a VelocImmune mouse immunized with infectious HCMV particles. Clone 1C10 neutralizes infection after virion binding to cells by targeting gH/gL envelope complexes and potently reduces infection of diverse HCMV strains in fibroblast, trophoblast, and epithelial cells. Antibody competition assays found that 1C10 recognizes a region of gH associated with broad neutralization and binds to soluble pentamer in the low nanomolar range. Importantly, 1C10 treatment significantly reduced virus proliferation in both fibroblast and epithelial cells. Further, the combination treatment of mAb 1C10 with ganciclovir reduced HCMV infection and proliferation in a synergistic manner. This work characterizes a neutralizing human mAb for potential use as a HCMV treatment, as well as a possible therapeutic strategy utilizing combination-based treatments targeting disparate steps of the viral life cycle. Collectively, the findings support an antibody-based therapy to effectively treat patients at risk for HCMV-associated diseases. IMPORTANCE: Human cytomegalovirus is a herpesvirus that infects a large proportion of the population and can cause significant disease in diverse patient populations whose immune systems are suppressed or compromised. The development and optimization of safe anti-HCMV therapeutics, especially those that have viral targets and inhibition mechanisms different from current HCMV treatments, are of urgent necessity to better public health. Human monoclonal antibodies (mAbs) that prevent HCMV entry of cells were identified by immunizing transgenic mice and screened for broad and effective neutralization capability. Here, we describe one such mAb, which was found to target gH/gL envelope complexes and effectively limit HCMV infection and dissemination. Further, administration of the antibody in combination with the antiviral drug ganciclovir inhibited HCMV in a synergistic manner, highlighting this approach and the use of anti-HCMV mAbs more broadly, as a potential therapeutic strategy for the treatment of diverse patient populations.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Antivirales , Infecciones por Citomegalovirus , Citomegalovirus , Ratones Transgénicos , Proteínas del Envoltorio Viral , Animales , Humanos , Citomegalovirus/inmunología , Citomegalovirus/efectos de los fármacos , Ratones , Anticuerpos Antivirales/inmunología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/prevención & control , Infecciones por Citomegalovirus/virología , Proteínas del Envoltorio Viral/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Virión/inmunología , Fibroblastos/virología , Replicación Viral/efectos de los fármacos , Anticuerpos ampliamente neutralizantes/inmunología , Antivirales/farmacología , Inmunización
2.
Clin Infect Dis ; 67(5): 770-777, 2018 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-29506084

RESUMEN

Background: Cytomegalovirus (CMV) has been linked to higher risk of cardiovascular disease and mortality. We aimed to determine if CMV is associated with neurocognitive performance in adults infected with human immunodeficiency virus (HIV). Methods: In this cross-sectional analysis, anti-CMV immunoglobulin G (IgG) concentrations in blood and CMV DNA copies in blood and cerebrospinal fluid (CSF) were measured in stored specimens of 80 HIV-infected adults who were previously assessed with a comprehensive neurocognitive test battery. Thirty-eight were taking suppressive antiretroviral therapy (ART) and 42 were not taking ART. A panel of 7 soluble biomarkers was measured by immunoassay in CSF. Results: Anti-CMV IgG concentrations ranged from 5.2 to 46.1 IU/mL. CMV DNA was detected in 7 (8.8%) plasma specimens but in no CSF specimens. Higher anti-CMV IgG levels were associated with older age (P = .0017), lower nadir CD4+ T-cell count (P < .001), AIDS (P < .001), and higher soluble CD163 (P = .009). Higher anti-CMV IgG levels trended toward an association with worse neurocognitive performance overall (P = .059). This correlation was only present in those taking suppressive ART (P = .0049). Worse neurocognitive performance remained associated with higher anti-CMV IgG levels after accounting for other covariates in multivariate models (model P = .0038). Detectable plasma CMV DNA was associated with AIDS (P = .05) but not with neurocognitive performance. Conclusions: CMV may influence neurocognitive performance in HIV-infected adults taking suppressive ART. Future clinical trials of anti-CMV therapy should help to determine whether the observed relationships are causal.


Asunto(s)
Anticuerpos Antivirales/sangre , Infecciones por Citomegalovirus/complicaciones , Infecciones por VIH/tratamiento farmacológico , Inmunoglobulina G/sangre , Trastornos Neurocognitivos/etiología , Adulto , Factores de Edad , Fármacos Anti-VIH/uso terapéutico , Terapia Antirretroviral Altamente Activa , Biomarcadores/líquido cefalorraquídeo , Estudios de Cohortes , Estudios Transversales , Citomegalovirus , Infecciones por Citomegalovirus/sangre , Infecciones por Citomegalovirus/inmunología , ADN Viral/sangre , ADN Viral/líquido cefalorraquídeo , Femenino , Humanos , Masculino , Pruebas de Estado Mental y Demencia , Trastornos Neurocognitivos/virología , Carga Viral
3.
J Virol ; 90(23): 10715-10727, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27654292

RESUMEN

Cytomegalovirus (CMV) is a ubiquitous human pathogen that increases the morbidity and mortality of immunocompromised individuals. The current FDA-approved treatments for CMV infection are intended to be virus specific, yet they have significant adverse side effects, including nephrotoxicity and hematological toxicity. Thus, there is a medical need for safer and more effective CMV therapeutics. Using a high-content screen, we identified the cardiac glycoside convallatoxin as an effective compound that inhibits CMV infection. Using a panel of cardiac glycoside variants, we assessed the structural elements critical for anti-CMV activity by both experimental and in silico methods. Analysis of the antiviral effects, toxicities, and pharmacodynamics of different variants of cardiac glycosides identified the mechanism of inhibition as reduction of methionine import, leading to decreased immediate-early gene translation without significant toxicity. Also, convallatoxin was found to dramatically reduce the proliferation of clinical CMV strains, implying that its mechanism of action is an effective strategy to block CMV dissemination. Our study has uncovered the mechanism and structural elements of convallatoxin, which are important for effectively inhibiting CMV infection by targeting the expression of immediate-early genes. IMPORTANCE: Cytomegalovirus is a highly prevalent virus capable of causing severe disease in certain populations. The current FDA-approved therapeutics all target the same stage of the viral life cycle and induce toxicity and viral resistance. We identified convallatoxin, a novel cell-targeting antiviral that inhibits CMV infection by decreasing the synthesis of viral proteins. At doses low enough for cells to tolerate, convallatoxin was able to inhibit primary isolates of CMV, including those resistant to the anti-CMV drug ganciclovir. In addition to identifying convallatoxin as a novel antiviral, limiting mRNA translation has a dramatic impact on CMV infection and proliferation.


Asunto(s)
Antivirales/farmacología , Infecciones por Citomegalovirus/prevención & control , Citomegalovirus/efectos de los fármacos , Metionina/metabolismo , Estrofantinas/farmacología , Antivirales/química , Transporte Biológico Activo/efectos de los fármacos , Glicósidos Cardíacos/química , Glicósidos Cardíacos/farmacología , Línea Celular , Citomegalovirus/genética , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/virología , Genes Inmediatos-Precoces/efectos de los fármacos , Genes Virales/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Estrofantinas/química , Relación Estructura-Actividad , Replicación Viral/efectos de los fármacos
4.
Virol J ; 12: 132, 2015 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-26319137

RESUMEN

BACKGROUND: Copper has antimicrobial properties and has been studied for its activity against viruses, including HIV. Copper complexed within a phthalocyanine ring, forming copper (II) phthalocyanine sulfate (CuPcS), may have a role in microbicide development when used intravaginally. METHODS: CuPcS toxicity was tested against cervical epithelial cells, TZM-BL cells, peripheral blood mononuclear cells (PBMC), and cervical explant tissues using cell viability assays. In vivo toxicity was assessed following intravaginal administration of CuPcS in female BALB/C mice and measured using a standardized histology grading system on reproductive tract tissues. Efficacy studies for preventing infection with HIV in the presence of various non-toxic concentrations of CuPcS were carried out in TZM-BL, PBMC, and cervical explant cultures using HIV-1BAL and various pseudovirus subtypes. Non-linear regression was applied to the data to determine the EC50/90 and CC50/90. RESULTS: CuPcS demonstrated inhibition of HIV infection in PBMCs at concentrations that were non-toxic in cervical epithelial cells and PBMCs with EC50 values of approximately 50 µg/mL. Reproductive tract tissue analysis revealed no toxicity at 100 mg/mL. Human cervical explant tissues challenged with HIV in the presence of CuPcS also revealed a dose-response effect at preventing HIV infection at non-toxic concentrations with an EC50 value of 65 µg/mL. CONCLUSION: These results suggest that CuPcS may be useful as a topical microbicide in concentrations that can be achieved in the female genital tract.


Asunto(s)
Antiinfecciosos Locales/farmacología , Transmisión de Enfermedad Infecciosa/prevención & control , Infecciones por VIH/prevención & control , Indoles/farmacología , Compuestos Organometálicos/farmacología , Sulfatos/farmacología , Administración Intravaginal , Animales , Antiinfecciosos Locales/efectos adversos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Infecciones por VIH/transmisión , Humanos , Indoles/efectos adversos , Ratones Endogámicos BALB C , Modelos Biológicos , Compuestos Organometálicos/efectos adversos , Sulfatos/efectos adversos , Resultado del Tratamiento
5.
J Immunol ; 191(2): 828-36, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23761635

RESUMEN

Lymphocyte activation is regulated by costimulatory and inhibitory receptors, of which both B and T lymphocyte attenuator (BTLA) and CD160 engage herpesvirus entry mediator (HVEM). Notably, it remains unclear how HVEM functions with each of its ligands during immune responses. In this study, we show that HVEM specifically activates CD160 on effector NK cells challenged with virus-infected cells. Human CD56(dim) NK cells were costimulated specifically by HVEM but not by other receptors that share the HVEM ligands LIGHT, Lymphotoxin-α, or BTLA. HVEM enhanced human NK cell activation by type I IFN and IL-2, resulting in increased IFN-γ and TNF-α secretion, and tumor cell-expressed HVEM activated CD160 in a human NK cell line, causing rapid hyperphosphorylation of serine kinases ERK1/2 and AKT and enhanced cytolysis of target cells. In contrast, HVEM activation of BTLA reduced cytolysis of target cells. Together, our results demonstrate that HVEM functions as a regulator of immune function that activates NK cells via CD160 and limits lymphocyte-induced inflammation via association with BTLA.


Asunto(s)
Antígenos CD/metabolismo , Células Asesinas Naturales/inmunología , Receptores Inmunológicos/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Antígeno CD56/metabolismo , Línea Celular , Activación Enzimática , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Humanos , Inflamación , Interferón Tipo I/metabolismo , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Células Asesinas Naturales/metabolismo , Activación de Linfocitos , Linfotoxina-alfa/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Transducción de Señal , Linfocitos T/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
6.
J Infect Dis ; 208(4): 564-72, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23661800

RESUMEN

Serum, cerebrospinal fluid (CSF), and cryopreserved lymphocytes from subjects in the Rush Alzheimer's Disease Center Religious Orders Study were analyzed for associations between cytomegalovirus (CMV) infection and clinical and pathological markers of Alzheimer disease. CMV antibody levels were associated with neurofibrillary tangles (NFTs). CSF interferon γ was only detected in seropositive subjects and was significantly associated with NFTs. The percentage of senescent T cells (CD4+ or CD8+CD28-CD57+) was significantly higher for CMV-seropositive as compared to CMV-seronegative subjects and was marginally associated with the pathologic diagnosis of Alzheimer disease (CD4+) or amyloid-ß (CD8+). Immunocytochemical analysis showed induction of amyloid-ß in human foreskin fibroblasts (HFFs) infected with each of 3 clinical CMV strains. In the same subjects, there was no association of herpes simplex virus type 1 (HSV-1) antibody levels with CMV antibody levels or clinical or pathological markers of Alzheimer disease. HSV-1 infection of HFFs did not induce amyloid-ß. These data support an association between CMV and the development of Alzheimer disease.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Anciano , Anciano de 80 o más Años , Péptidos beta-Amiloides/análisis , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/líquido cefalorraquídeo , Infecciones por Citomegalovirus/complicaciones , Femenino , Fibroblastos/química , Fibroblastos/virología , Humanos , Interferón gamma/líquido cefalorraquídeo , Masculino , Ovillos Neurofibrilares/metabolismo , Subgrupos de Linfocitos T/inmunología
7.
J Infect Dis ; 205(12): 1788-96, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22492856

RESUMEN

BACKGROUND: Cytomegalovirus (CMV) infection has been implicated in immune activation and accelerated progression of immunodeficiency from human immunodeficiency virus (HIV) coinfection. We hypothesized that CMV is associated with vascular disease in HIV-infected adults. METHODS: In the Women's Interagency HIV Study, we studied 601 HIV-infected and 90 HIV-uninfected participants. We assessed the association of CMV immunoglobulin G (IgG) level with carotid artery intima-media thickness, carotid artery distensibility, Young's elastic modulus, and blood pressures. Multivariable models adjusted for age, race/ethnicity, smoking, diabetes, and body mass index. RESULTS: Mean CMV IgG levels were higher in HIV-infected women compared with HIV-uninfected women (P < .01). Among HIV-infected women, higher CMV IgG level was associated with decreased carotid artery distensibility (P < .01) and increased Young's modulus (P = .02). Higher CMV IgG antibody level was associated with increased prevalence of carotid artery lesions among HIV-infected women who achieved HIV suppression on antiretroviral therapy, but not among viremic or untreated HIV-infected women. Adjustment for Epstein-Barr virus antibody levels and C-reactive protein levels had no effect on the associations between CMV IgG levels and vascular parameters. CONCLUSIONS: Cytomegalovirus antibody titers are increased in HIV-infected women and associated with subclinical cardiovascular disease. Host responses to CMV may be abnormal in HIV infection and associated with clinical disease.


Asunto(s)
Anticuerpos Antivirales/inmunología , Enfermedades de las Arterias Carótidas/etiología , Infecciones por Citomegalovirus/epidemiología , Infecciones por Citomegalovirus/inmunología , Citomegalovirus/inmunología , Infecciones por VIH/complicaciones , Inmunoglobulina G/inmunología , Adulto , Arterias Carótidas/patología , Femenino , Humanos , Persona de Mediana Edad , Prevalencia , Túnica Íntima/patología
8.
J Virol ; 85(24): 13204-13, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21976654

RESUMEN

Human cytomegalovirus (HCMV) is the leading cause of congenital infection, associated with severe birth defects and intrauterine growth retardation. The mechanism of HCMV transmission via the maternal-fetal interface is largely unknown, and there are no animal models for HCMV. The initial stages of infection are believed to occur in the maternal decidua. Here we employed a novel decidual organ culture, using both clinically derived and laboratory-derived viral strains, for the ex vivo modeling of HCMV transmission in the maternal-fetal interface. Viral spread in the tissue was demonstrated by the progression of infected-cell foci, with a 1.3- to 2-log increase in HCMV DNA and RNA levels between days 2 and 9 postinfection, the expression of immediate-early and late proteins, the appearance of typical histopathological features of natural infection, and dose-dependent inhibition of infection by ganciclovir and acyclovir. HCMV infected a wide range of cells in the decidua, including invasive cytotrophoblasts, macrophages, and endothelial, decidual, and dendritic cells. Cell-to-cell viral spread was revealed by focal extension of infected-cell clusters, inability to recover infectious extracellular virus, and high relative proportions (88 to 93%) of cell-associated viral DNA. Intriguingly, neutralizing HCMV hyperimmune globulins exhibited inhibitory activity against viral spread in the decidua even when added at 24 h postinfection-providing a mechanistic basis for their clinical use in prenatal prevention. The ex vivo-infected decidual cultures offer unique insight into patterns of viral tropism and spread, defining initial stages of congenital HCMV transmission, and can facilitate evaluation of the effects of new antiviral interventions within the maternal-fetal interface milieu.


Asunto(s)
Infecciones por Citomegalovirus/transmisión , Decidua/virología , Transmisión Vertical de Enfermedad Infecciosa , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , ADN Viral/genética , ADN Viral/aislamiento & purificación , Femenino , Expresión Génica , Humanos , Modelos Biológicos , Técnicas de Cultivo de Órganos/métodos , Embarazo , ARN Viral/genética , ARN Viral/aislamiento & purificación , Factores de Tiempo , Proteínas Virales/biosíntesis
9.
Clin Microbiol Rev ; 23(4): 689-712, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20930070

RESUMEN

The study of human cytomegalovirus (HCMV) antiviral drug resistance has enhanced knowledge of the virological targets and the mechanisms of antiviral activity. The currently approved drugs, ganciclovir (GCV), foscarnet (FOS), and cidofovir (CDV), target the viral DNA polymerase. GCV anabolism also requires phosphorylation by the virus-encoded UL97 kinase. GCV resistance mutations have been identified in both genes, while FOS and CDV mutations occur only in the DNA polymerase gene. Confirmation of resistance mutations requires phenotypic analysis; however, phenotypic assays are too time-consuming for diagnostic purposes. Genotypic assays based on sequencing provide more rapid results but are dependent on prior validation by phenotypic methods. Reports from many laboratories have produced an evolving list of confirmed resistance mutations, although differences in interpretation have led to some confusion. Recombinant phenotyping methods performed in a few research laboratories have resolved some of the conflicting results. Treatment options for drug-resistant HCMV infections are complex and have not been subjected to controlled clinical trials, although consensus guidelines have been proposed. This review summarizes the virological and clinical data pertaining to HCMV antiviral drug resistance.


Asunto(s)
Antivirales/farmacología , Citomegalovirus/efectos de los fármacos , Farmacorresistencia Viral/genética , Antivirales/uso terapéutico , Cidofovir , Citomegalovirus/genética , Infecciones por Citomegalovirus/tratamiento farmacológico , Citosina/análogos & derivados , Citosina/farmacología , Citosina/uso terapéutico , ADN Polimerasa Dirigida por ADN , Foscarnet/farmacología , Foscarnet/uso terapéutico , Ganciclovir/farmacología , Ganciclovir/uso terapéutico , Genoma Viral , Humanos , Organofosfonatos/farmacología , Organofosfonatos/uso terapéutico , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Análisis de Secuencia de ADN
10.
J Infect Dis ; 203(4): 452-63, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21220772

RESUMEN

BACKGROUND: Individuals infected with human immunodeficiency virus (HIV) have increased risk of cardiovascular events. It is unknown whether T cell activation and senescence, 2 immunologic sequelae of HIV infection, are associated with vascular disease among HIV-infected adults. METHODS: T cell phenotyping and carotid ultrasound were assessed among 115 HIV-infected women and 43 age- and race/ethnicity-matched HIV-uninfected controls participating in the Women's Interagency HIV Study. Multivariate analyses were used to assess the association of T cell activation (CD38(+)HLA-DR(+)) and senescence (CD28(-)CD57(+)) with subclinical carotid artery disease. RESULTS: Compared with HIV-uninfected women, frequencies of CD4(+)CD38(+)HLA-DR(+), CD8(+)CD38(+)HLA-DR(+), and CD8(+)CD28(-)CD57(+) T cells were higher among HIV-infected women, including those who achieved viral suppression while receiving antiretroviral treatment. Among HIV-infected women, adjusted for age, antiretroviral medications, and viral load, higher frequencies of activated CD4(+) and CD8(+) T cells and immunosenescent CD8(+) T cells were associated with increased prevalence of carotid artery lesions (prevalence ratio(lesions) associated with activated CD4(+) T cells, 1.6 per SD [95% confidence interval {CI}, 1.1-2.2]; P = .02; prevalence ratio(lesions) associated with activated CD8(+) T cells, 2.0 per SD [95% CI, 1.2-3.3]; P < .01; prevalence ratio(lesions) associated with senescent CD8(+) T cells, 1.9 per SD [95% CI, 1.1-3.1]; P = .01). CONCLUSIONS: HIV-associated T cell changes are associated with subclinical carotid artery abnormalities, which may be observed even among those patients achieving viral suppression with effective antiretroviral therapy.


Asunto(s)
Enfermedades Asintomáticas/epidemiología , Enfermedades de las Arterias Carótidas/epidemiología , Infecciones por VIH/complicaciones , Infecciones por VIH/inmunología , Subgrupos Linfocitarios/inmunología , Linfocitos T/inmunología , ADP-Ribosil Ciclasa 1/análisis , Adulto , Antígenos CD28/análisis , Antígenos CD57/análisis , Arterias Carótidas/diagnóstico por imagen , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/diagnóstico , Femenino , Proteínas Fetales , Antígenos HLA-DR/análisis , Humanos , Subgrupos Linfocitarios/química , Persona de Mediana Edad , Proteínas de Dominio T Box , Linfocitos T/química , Ultrasonografía
11.
J Clin Microbiol ; 49(8): 2854-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21697326

RESUMEN

The performance characteristics of four different assays for hepatitis B virus (HBV) quantification were assessed: the Abbott RealTime HBV IUO, the Roche Cobas AmpliPrep/Cobas TaqMan HBV test, the Roche Cobas TaqMan HBV test with HighPure system, and the Qiagen artus HBV TM ASR. Limit of detection (LOD), linear range, reproducibility, and agreement were determined using a serially diluted plasma sample from a single chronically infected subject. Each assay was tested by at least three laboratories. The LOD of the RealTime and two TaqMan assays was approximately 1.0 log(10) IU/ml; for artus HBV (which used the lowest volume of extracted DNA), it was approximately 1.5 log(10) IU/ml. The linear range spanned 1.0 to at least 7.0 log(10) IU/ml for all assays. Median values were consistently lowest for artus HBV and highest for Cobas AmpliPrep/Cobas TaqMan HBV. Assays incorporating automated nucleic acid extraction were the most reproducible; however, the overall variability was minor since the standard deviations for the means of all tested concentrations were ≤0.32 log(10) IU/ml for all assays. False-positive results were observed with all assays; the highest rates occurred with tests using manual nucleic acid extraction. The performance characteristics of these assays suggest that they are useful for management and therapeutic monitoring of chronic HBV infection.


Asunto(s)
ADN Viral/aislamiento & purificación , Virus de la Hepatitis B/aislamiento & purificación , Hepatitis B/virología , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Carga Viral/métodos , ADN Viral/genética , Virus de la Hepatitis B/genética , Humanos , Plasma/virología , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
13.
Sci Rep ; 10(1): 7499, 2020 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-32372013

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

14.
AIDS ; 34(6): 849-857, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32271250

RESUMEN

BACKGROUND: Even with antiretroviral therapy (ART), persons with HIV (PWH) experience increased morbidity and mortality. Cytomegalovirus (CMV) and Epstein--Barr virus (EBV) co-infections likely exacerbate inflammatory-related diseases. OBJECTIVE: To determine if presence of detectable CMV or EBV DNA in peripheral blood mononuclear cells (PBMC) is associated with non-AIDS events among PWH receiving modern ART. DESIGN: We performed a case--control study of PWH starting ART and HIV-suppressed at year 1 and thereafter, 140 cases who experienced non-AIDS events and 305 matched controls. Events included myocardial infarction, stroke, malignancy, serious bacterial infection or death. METHODS: Blood samples were studied pre-ART, 1-year post-ART and pre-event. Controls had an event-free follow-up equal or greater than cases. CMV and EBV DNA levels were measured in PBMC. Conditional logistic regression analysis assessed associations and adjusted for relevant covariates; Spearman's correlations compared CMV and EBV DNA levels with other biomarkers. RESULTS: CMV DNA was detected in PBMC of 25% of participants, EBV DNA was detected in more than 90%. Higher EBV DNA levels were associated with increased risk of events at all time points (odds ratio (OR) per one IQR = 1.5-1.7, all P < 0.009). At year 1, detectable CMV DNA was associated with increased risk of events in most adjusted models (OR = 1.4-1.8, P values ranging 0.03-0.17). Higher levels of CMV and EBV DNA correlated with multiple inflammatory markers and lower CD4/CD8 ratio. CONCLUSION: In PWH starting ART, detection of CMV and EBV DNA in PBMC was associated with development of non-AIDS events. Clinical trials will be needed to understand causal mechanisms and ways to interrupt them.


Asunto(s)
Infecciones por Citomegalovirus/sangre , Citomegalovirus/aislamiento & purificación , ADN Viral/genética , Infecciones por Virus de Epstein-Barr/sangre , Herpesvirus Humano 4/aislamiento & purificación , Adulto , Terapia Antirretroviral Altamente Activa , Estudios de Casos y Controles , Citomegalovirus/genética , Infecciones por Citomegalovirus/complicaciones , ADN Viral/metabolismo , Infecciones por Virus de Epstein-Barr/complicaciones , Femenino , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Herpesvirus Humano 4/genética , Humanos , Leucocitos Mononucleares , Masculino , Persona de Mediana Edad
15.
Sci Rep ; 10(1): 4746, 2020 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-32179788

RESUMEN

Ginkgolic acids (GA) are alkylphenol constituents of the leaves and fruits of Ginkgo biloba. GA has shown pleiotropic effects in vitro, including: antitumor effects through inhibition of lipogenesis; decreased expression of invasion associated proteins through AMPK activation; and potential rescue of amyloid-ß (Aß) induced synaptic impairment. GA was also reported to have activity against Escherichia coli and Staphylococcus aureus. Several mechanisms for this activity have been suggested including: SUMOylation inhibition; blocking formation of the E1-SUMO intermediate; inhibition of fatty acid synthase; non-specific SIRT inhibition; and activation of protein phosphatase type-2C. Here we report that GA inhibits Herpes simplex virus type 1 (HSV-1) by inhibition of both fusion and viral protein synthesis. Additionally, we report that GA inhibits human cytomegalovirus (HCMV) genome replication and Zika virus (ZIKV) infection of normal human astrocytes (NHA). We show a broad spectrum of fusion inhibition by GA of all three classes of fusion proteins including HIV, Ebola virus (EBOV), influenza A virus (IAV) and Epstein Barr virus (EBV). In addition, we show inhibition of a non-enveloped adenovirus. Our experiments suggest that GA inhibits virion entry by blocking the initial fusion event. Data showing inhibition of HSV-1 and CMV replication, when GA is administered post-infection, suggest a possible secondary mechanism targeting protein and DNA synthesis. Thus, in light of the strong effect of GA on viral infection, even after the infection begins, it may potentially be used to treat acute infections (e.g. Coronavirus, EBOV, ZIKV, IAV and measles), and also topically for the successful treatment of active lesions (e.g. HSV-1, HSV-2 and varicella-zoster virus (VZV)).


Asunto(s)
Antivirales/farmacología , Infecciones por Virus ADN/metabolismo , Virus ADN/efectos de los fármacos , Infecciones por Virus ARN/metabolismo , Virus ARN/efectos de los fármacos , Salicilatos/farmacología , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Proteínas Virales de Fusión/antagonistas & inhibidores , Animales , Astrocitos/metabolismo , Chlorocebus aethiops , Replicación del ADN/efectos de los fármacos , Infecciones por Virus ADN/virología , Virus ADN/genética , ADN Viral/genética , Células HEK293 , Humanos , Infecciones por Virus ARN/virología , Virus ARN/genética , Células Vero , Proteínas del Envoltorio Viral/biosíntesis , Proteínas Virales de Fusión/biosíntesis , Virión/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
16.
J Gen Virol ; 90(Pt 10): 2375-2380, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19553388

RESUMEN

The genomes of commonly used variants of human cytomegalovirus (HCMV) strains Towne and AD169 each contain a substantial mutation in which a region (U(L)/b') at the right end of the long unique region has been replaced by an inverted duplication of a region from the left end of the genome. Using high-throughput technology, we have sequenced HCMV strain Towne (ATCC VR-977) and confirmed the presence of two variants, one exhibiting the replacement in U(L)/b' and the other intact in this region. Both variants are mutated in genes RL13, UL1, UL40, UL130, US1 and US9. We have also sequenced a novel AD169 variant (varUC) that is intact in U(L)/b' except for a small deletion that affects genes UL144, UL142, UL141 and UL140. Like other AD169 variants, varUC is mutated in genes RL5A, RL13, UL36 and UL131A. A subpopulation of varUC contains an additional deletion affecting genes IRS1, US1 and US2.


Asunto(s)
Citomegalovirus/clasificación , Citomegalovirus/genética , Variación Genética , Secuencia de Bases , ADN Viral , Genoma Viral , Humanos , Mutación
17.
J Clin Microbiol ; 47(11): 3530-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19726602

RESUMEN

Microbicide candidates with promising in vitro activity are often advanced for evaluations using human primary tissue explants relevant to the in vivo mucosal transmission of human immunodeficiency virus type 1 (HIV-1), such as tonsil, cervical, or rectal tissue. To compare virus growth or the anti-HIV-1 efficacies of candidate microbicides in tissue explants, a novel soft-endpoint method was evaluated to provide a single, objective measurement of virus growth. The applicability of the soft endpoint is shown across several different ex vivo tissue types, with the method performed in different laboratories, and for a candidate microbicide (PRO 2000). The soft-endpoint method was compared to several other endpoint methods, including (i) the growth of virus on specific days after infection, (ii) the area under the virus growth curve, and (iii) the slope of the virus growth curve. Virus growth at the assay soft endpoint was compared between laboratories, methods, and experimental conditions, using nonparametric statistical analyses. Intra-assay variability determinations using the coefficient of variation demonstrated higher variability for virus growth in rectal explants. Significant virus inhibition by PRO 2000 and significant differences in the growth of certain primary HIV-1 isolates were observed by the majority of laboratories. These studies indicate that different laboratories can provide consistent measurements of anti-HIV-1 microbicide efficacy when (i) the soft endpoint or another standardized endpoint is used, (ii) drugs and/or virus reagents are centrally sourced, and (iii) the same explant tissue type and method are used. Application of the soft-endpoint method reduces the inherent variability in comparisons of preclinical assays used for microbicide development.


Asunto(s)
Antiinfecciosos/farmacología , VIH-1/efectos de los fármacos , Pruebas de Sensibilidad Microbiana/métodos , Pruebas de Sensibilidad Microbiana/normas , Cuello del Útero/virología , Femenino , Humanos , Técnicas In Vitro , Masculino , Membrana Mucosa/virología , Tonsila Palatina/virología , Recto/virología , Reproducibilidad de los Resultados , Replicación Viral/efectos de los fármacos
18.
Pathog Immun ; 4(1): 66-78, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30815626

RESUMEN

BACKGROUND: Among HIV-positive individuals, increased levels of inflammation and immune activation persist even in the setting of effective antiretroviral therapy (ART) and are associated with greater rates of non-AIDS events. The etiology of this persistent inflammation is incompletely understood. METHODS: Using a well-characterized cohort of 322 HIV-infected individuals on suppressive ART, we conducted a case-control study. Cytomegalovirus (CMV) immunoglobulin G (IgG) levels, plasma biomarkers, and T-cell phenotypes were measured/characterized from samples collected 1 year after ART initiation. Conditional logistic regression for matched case-control studies analyzed the associations of year 1 CMV-specific IgG level with the subsequent occurrence of any non-AIDS event. Correlations between continuous CMV IgG antibody levels and soluble and cellular markers were assessed. RESULTS: We found that higher levels of CMV IgG were associated with increased risk of non-AIDS events (OR = 1.58 per IQR [95% CI: 1.12, 2.24], P = 0.01) and with elevated soluble and cellular markers of inflammation. CONCLUSIONS: The magnitude of the host immune response to CMV may play a role in the persistent inflammation and resultant morbid events observed in the HIV-positive population.

19.
Clin Infect Dis ; 46(9): 1455-7, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18419454

RESUMEN

This is the first report of treatment of cytomegalovirus infection with artesunate, for a stem cell transplant recipient with a newly identified foscarnet-resistant and ganciclovir-resistant DNA polymerase L776M mutation. Artesunate treatment resulted in a 1.7-2.1-log reduction in viral load by treatment day 7, with a viral half-life of 0.9-1.9 days, indicating a highly effective block in viral replication.


Asunto(s)
Artemisininas/uso terapéutico , Infecciones por Citomegalovirus/tratamiento farmacológico , Trasplante de Células Madre Hematopoyéticas/métodos , Antivirales/farmacología , Antivirales/uso terapéutico , Artemisininas/farmacología , Artesunato , Niño , Citomegalovirus/efectos de los fármacos , Infecciones por Citomegalovirus/etiología , Farmacorresistencia Viral , Foscarnet/farmacología , Foscarnet/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Masculino , Resultado del Tratamiento
20.
AIDS Res Hum Retroviruses ; 24(3): 453-62, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18327985

RESUMEN

HIV-1 copathogens are believed to play a critical role in progression to AIDS. Human cytomegalovirus (HCMV) has a high prevalence in the general population and is a common copathogen in HIV-1-infected individuals. Important events in copathogen interactions with HIV-1 take place in lymphoid tissue where critical events in HIV-1 disease occur. Here, we used an experimental system of human lymphoid tissue ex vivo to investigate interactions of HCMV with HIV-1. We inoculated ex vivo blocks of human lymphoid tissue with a recombinant strain of HCMV, expressing the green fluorescent protein, and HIV-1 and monitored viral replication and the phenotype of productively infected cells. HCMV readily replicated in tissue blocks as revealed by the release of HCMV viral DNA and an increasing number of viral-positive cells. Immunophenotyping of HCMV-infected cells showed a preferential infection of activated lymphocytes. The number of these cells significantly increased in HIV-1-coinfected tissues. Accordingly, HCMV replication was enhanced 2- to-3 fold. This upregulation occurred in tissues infected with either CXCR4- or CCR5-utilizing HIV-1. Thus, HIV-1 creates new targets for HCMV, which may explain the strong association of HCMV with HIV-1 infection in vivo. Ex vivo-infected human lymphoid tissue constitutes a model to study the mechanisms of HCMV tissue pathogenesis and its interactions with HIV-1 and this model may provide new targets for anti-HIV-1 therapy.


Asunto(s)
Citomegalovirus/crecimiento & desarrollo , VIH-1/crecimiento & desarrollo , Tejido Linfoide/virología , Replicación Viral , Citomegalovirus/genética , ADN Viral/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Linfocitos/virología , Técnicas de Cultivo de Órganos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA