Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Microbiol Spectr ; 9(3): e0102421, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34908456

RESUMEN

Hepatitis D is the most severe form of human viral hepatitis and currently lacks an efficient therapy. Dendritic cell-derived exosomes (Dexs) have been found to induce immune responses capable of eliminating viruses. However, the therapeutic potential of antigen-loaded exosomes in hepatitis D is still unknown. Recently, we designed exosomes loaded with ubiquitinated hepatitis delta virus (HDV) small delta antigen (Ub-S-HDAg) and then treated mice bearing replicating HDV with these exosomes to explore their antiviral effect and mechanism. Mature dendritic cell-derived exosomes (mDexs) were loaded with Ub-S-HDAg and their antivirus function was evaluated in mice with HDV viremia. Furthermore, the proportion of CD8+ cells, the ratio of Th1/Th2 cells, the postimmunization levels of cytokines were explored, and the Janus kinases (JAK)/signal transducer and activator of transcription (STAT) pathway was evaluated with a JAK2 inhibitor AG490. In Ub-S-HDAg-Dexs group, the HDV RNA viral load was significantly decreased compared with other groups by CD8+ cell enrichment and an increase Th1/Th2 cell ratio. Furthermore, lymphocyte infiltration was increased, while the HDAg level was decreased in mouse liver tissue. However, there were no significant differences in HBV surface antigen (HBsAg), alanine aminotransferase (ALT), or aspartate aminotransferase (AST) levels among the groups. Moreover, p-JAK2, p-STAT1, p-STAT4, STAT1, and STAT4 expression was increased in Ub-S-HDAg-Dexs group. In conclusion, Ub-S-HDAg-Dexs might be a potential immunotherapeutic agent for eradicating HDV by inducing specific cellular immune response via the JAK/STAT pathway. IMPORTANCE Hepatitis D is the most severe viral hepatitis with accelerating the process of liver cirrhosis and increasing the risk of hepatocellular carcinoma. However, there are no effective antiviral drugs. Exosomes derived from mature dendritic cells are used not only as immunomodulators, but also as biological carriers to deliver antigens to induce robust immune response. Based on these properties, exosomes could be used as a biological immunotherapy by enhancing adaptive immune response to inhibit hepatitis D virus replication. Our research may provide a new therapeutic strategy to eradicate HDV in the future.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Micropartículas Derivadas de Células/inmunología , Exosomas/inmunología , Virus de la Hepatitis Delta/inmunología , Antígenos de Hepatitis delta/inmunología , Balance Th1 - Th2/fisiología , Alanina Transaminasa/análisis , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Aspartato Aminotransferasas/análisis , Micropartículas Derivadas de Células/virología , Células Cultivadas , Citocinas/sangre , Células Dendríticas/inmunología , Exosomas/virología , Femenino , Antígenos de Superficie de la Hepatitis B/análisis , Antígenos de Hepatitis delta/metabolismo , Factores Inmunológicos/farmacología , Inmunoterapia/métodos , Janus Quinasa 2/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Inhibidores de Proteínas Quinasas/farmacología , Tirfostinos/farmacología , Carga Viral , Replicación Viral/inmunología
2.
Commun Biol ; 4(1): 652, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34079056

RESUMEN

Hantaan viruses (HTNVs) are zoonotic pathogens transmitted mainly by rodents and capable of infecting humans. Increasing knowledge of the human response to HTNV infection can guide the development of new preventative vaccines and therapeutic strategies. Here, we show that HTNV can infect CD8+ T cells in vivo in patients diagnosed with hemorrhagic fever with renal syndrome (HFRS). Electron microscopy-mediated tracking of the life cycle and ultrastructure of HTNV-infected CD8+ T cells in vitro showed an association between notable increases in cytoplasmic multivesicular bodies and virus production. Notably, based on a clinical cohort of 280 patients, we found that circulating HTNV-infected CD8+ T cell numbers in blood were proportional to disease severity. These results demonstrate that viral infected CD8+ T cells may be used as an adjunct marker for monitoring HFRS disease progression and that modulating T cell functions may be explored for new treatment strategies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Virus Hantaan/inmunología , Virus Hantaan/patogenicidad , Fiebre Hemorrágica con Síndrome Renal/inmunología , Fiebre Hemorrágica con Síndrome Renal/virología , Enfermedad Aguda , Adulto , Linfocitos T CD8-positivos/ultraestructura , Micropartículas Derivadas de Células/ultraestructura , Micropartículas Derivadas de Células/virología , Citocinas/sangre , Progresión de la Enfermedad , Femenino , Virus Hantaan/fisiología , Fiebre Hemorrágica con Síndrome Renal/sangre , Humanos , Técnicas In Vitro , Masculino , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Modelos Biológicos , Virión/inmunología , Virión/patogenicidad , Replicación Viral
3.
J Am Heart Assoc ; 9(17): e015998, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32819189

RESUMEN

Background Microvesicles are cell membrane-derived vesicles that have been shown to augment inflammation. Specifically, monocyte-derived microvesicles (MDMVs), which can express the coagulation protein tissue factor, contribute to thrombus formation and cardiovascular disease. People living with HIV experience higher prevalence of cardiovascular disease and also exhibit increased levels of plasma microvesicles. The process of microvesicle release has striking similarity to budding of enveloped viruses. The surface protein tetherin inhibits viral budding by physically tethering budding virus particles to cells. Hence, we investigated the role of tetherin in regulating the release of MDMVs during HIV infection. Methods and Results The plasma of aviremic HIV-infected individuals had increased levels of tissue factor + MDMVs, as measured by flow cytometry, and correlated to reduced tetherin expression on monocytes. Superresolution confocal and electron microscopy showed that tetherin localized at the site of budding MDMVs. Mechanistic studies revealed that the exposure of monocytes to HIV-encoded Tat triggered tetherin loss and subsequent rise in MDMV production. Overexpression of tetherin in monocytes led to morphologic changes in the pseudopodia directly underneath the MDMVs. Further, tetherin knockout mice demonstrated a higher number of circulating MDMVs and less time to bleeding cessation. Conclusions Our studies define a novel regulatory mechanism of MDMV release through tetherin and explore its contribution to the procoagulatory state that is frequently observed in people with HIV. Such insights could lead to improved therapies for individuals infected with HIV and also for those with cardiovascular disease.


Asunto(s)
Antivirales/metabolismo , Antígeno 2 del Estroma de la Médula Ósea/metabolismo , Micropartículas Derivadas de Células/genética , Infecciones por VIH/metabolismo , Adulto , Animales , Factores de Coagulación Sanguínea/metabolismo , Antígeno 2 del Estroma de la Médula Ósea/farmacología , Antígeno 2 del Estroma de la Médula Ósea/ultraestructura , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/etiología , Membrana Celular/metabolismo , Micropartículas Derivadas de Células/patología , Micropartículas Derivadas de Células/virología , Femenino , VIH/efectos de los fármacos , Infecciones por VIH/sangre , Infecciones por VIH/complicaciones , Infecciones por VIH/virología , Humanos , Inmunohistoquímica/métodos , Inflamación/metabolismo , Receptores de Lipopolisacáridos/metabolismo , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Monocitos/metabolismo , Prevalencia , Proteínas Reguladoras y Accesorias Virales/metabolismo
4.
Neurology ; 94(5): e474-e480, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31892634

RESUMEN

OBJECTIVE: Varicella zoster virus (VZV) can spread anterogradely and infect cerebral arteries causing VZV vasculopathy and arterial ischemic stroke. In this study, we tested the hypothesis that virus-infected cerebrovascular fibroblasts undergo phenotypic changes that promote vascular remodeling and facilitate virus transmission in an in vitro model of VZV vasculopathy. The aims of this project were therefore to examine the changes that virus-infected human brain adventitial vascular fibroblasts (HBVAFs) undergo in an in vitro model of VZV vasculopathy and to identify disease biomarkers relating to VZV-related vasculopathy. METHODS: HBVAFs were infected with VZV, and their ability to migrate, proliferate, transdifferentiate, and interact with endothelial cells was studied with flow cytometry. Microparticles (MPs) released from these cells were isolated and imaged with transmission electron microscopy, and their protein content was analyzed with mass spectrometry. Circulating MP profiles were also studied in children with VZV and non-VZV vasculopathy and compared with controls. RESULTS: VZV-infected HBVAFs transdifferentiated into myofibroblasts with enhanced proliferative and migratory capacity. Interaction of VZV-infected HBVAFs with endothelial cells resulted in endothelial dysfunction. These effects were, in part, mediated by the release of MPs from VZV-infected HBVAFs. These MPs contained VZV virions that could transmit VZV to neighboring cells, highlighting a novel model of VZV cell-to-cell viral dissemination. MPs positive for VZV were significantly higher in children with VZV-related vasculopathy compared to children with non-VZV vasculopathy (p = 0.01) and controls (p = 0.007). CONCLUSIONS: VZV-infected HBVAFs promote vascular remodeling and facilitate virus transmission. These effects were mediated by the release of apoptotic MPs that could transmit VZV infection to neighboring cells through a Trojan horse means of productive viral infection. VZV+ MPs may represent a disease biomarker worthy of further study.


Asunto(s)
Movimiento Celular , Proliferación Celular , Transdiferenciación Celular , Micropartículas Derivadas de Células/virología , Trastornos Cerebrovasculares/virología , Fibroblastos/virología , Miofibroblastos/virología , Remodelación Vascular , Adolescente , Adventicia , Micropartículas Derivadas de Células/ultraestructura , Arterias Cerebrales , Trastornos Cerebrovasculares/fisiopatología , Niño , Preescolar , Células Endoteliales , Femenino , Fibroblastos/fisiología , Fibroblastos/ultraestructura , Herpesvirus Humano 3 , Humanos , Técnicas In Vitro , Masculino , Espectrometría de Masas , Microscopía Electrónica de Transmisión , Miofibroblastos/fisiología , Miofibroblastos/ultraestructura , Accidente Cerebrovascular/fisiopatología , Accidente Cerebrovascular/virología , Cultivo de Virus
5.
Methods Mol Biol ; 2060: 305-317, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31617186

RESUMEN

Extracellular vesicles (EVs) are secreted membrane vesicles, derived from endosomes or from the plasma membrane, which have been isolated from most cell types and biological fluids. Although EVs are highly heterogeneous and their classification is complex, two major categories can be distinguished: microvesicles (MVs), which derive from the shedding of the plasma membrane, and exosomes, which correspond to intraluminal vesicles released to the extracellular milieu upon fusion of multivesicular bodies (MVBs) with the plasma membrane. Cells infected with viruses may secrete MVs containing viral proteins, RNAs and, in some instances, infectious virions. A recent study carried out by our laboratory has shown that MVs released by cells infected with HSV-1 contained virions and were endocytosed by naïve cells leading to a productive infection. This suggests that HSV-1 may use MVs for spreading, expanding its tropism and evading the host immune response. Here we describe in detail the methods used to isolate and analyse the MVs released from HSV-1-infected cells.


Asunto(s)
Micropartículas Derivadas de Células , Herpes Simple/metabolismo , Herpesvirus Humano 1 , Línea Celular , Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/virología , Herpes Simple/patología , Herpesvirus Humano 1/química , Herpesvirus Humano 1/aislamiento & purificación , Herpesvirus Humano 1/metabolismo , Humanos
6.
Virchows Arch ; 475(2): 175-180, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31203443

RESUMEN

Expression of the latent membrane protein-1 (LMP1) of Epstein-Barr virus (EBV) was investigated in 153 cases of EBV+ classic Hodgkin lymphoma (cHL); 120 cases were pediatric patients (< 14 years of age) from Iraq, and 33 cases were adult patients from Italy. We describe for the first time the presence of LMP1 protein in EBV-encoded RNA (EBER)-negative follicular dendritic cells (FDCs) of reactive germinal centers (GC) associated with EBV+ cHL. Presence of LMP1+ GCs was independent of geographic region and age of patients. Variable numbers of reactive GCs were present in 22.2% of cases (34 of 153), whereas LMP1 staining of FDCs was present in about a third of cases (10 of 34) with reactive GC. Most cases with LMP1+ GC were mixed-cellularity (MC) subtype, but some nodular sclerosis (NS) was also present. GC cells with LMP1+ FDCs were surrounded by numerous EBV-infected cells which were positive for EBER, LMP1, and CD30. Double immunolocalization analysis revealed that LMP1 was associated with CD63, an exosomal marker, and with CD21. The possibility is discussed that peri-follicular EBV-infected cells release LMP1 protein, perhaps through exosomes, and that the protein is then captured by FDCs and is presented to EBER-negative GC B cells.


Asunto(s)
Células Dendríticas Foliculares/virología , Infecciones por Virus de Epstein-Barr/virología , Enfermedad de Hodgkin/virología , Proteínas de la Matriz Viral/metabolismo , Adulto , Anciano , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/virología , Niño , Femenino , Centro Germinal/virología , Enfermedad de Hodgkin/patología , Humanos , Masculino , Persona de Mediana Edad
7.
J Virol ; 92(10)2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29514899

RESUMEN

Herpes simplex virus 1 (HSV-1) is a neurotropic pathogen that can infect many types of cells and establishes latent infections in the neurons of sensory ganglia. In some cases, the virus spreads into the central nervous system, causing encephalitis or meningitis. Cells infected with several different types of viruses may secrete microvesicles (MVs) containing viral proteins and RNAs. In some instances, extracellular microvesicles harboring infectious virus have been found. Here we describe the features of shedding microvesicles released by the human oligodendroglial HOG cell line infected with HSV-1 and their participation in the viral cycle. Using transmission electron microscopy, we detected for the first time microvesicles containing HSV-1 virions. Interestingly, the Chinese hamster ovary (CHO) cell line, which is resistant to infection by free HSV-1 virions, was susceptible to HSV-1 infection after being exposed to virus-containing microvesicles. Therefore, our results indicate for the first time that MVs released by infected cells contain virions, are endocytosed by naive cells, and lead to a productive infection. Furthermore, infection of CHO cells was not completely neutralized when virus-containing microvesicles were preincubated with neutralizing anti-HSV-1 antibodies. The lack of complete neutralization and the ability of MVs to infect nectin-1/HVEM-negative CHO-K1 cells suggest a novel way for HSV-1 to spread to and enter target cells. Taken together, our results suggest that HSV-1 could spread through microvesicles to expand its tropism and that microvesicles could shield the virus from neutralizing antibodies as a possible mechanism to escape the host immune response.IMPORTANCE Herpes simplex virus 1 (HSV-1) is a neurotropic pathogen that can infect many types of cells and establishes latent infections in neurons. Extracellular vesicles are a heterogeneous group of membrane vesicles secreted by most cell types. Microvesicles, which are extracellular vesicles which derive from the shedding of the plasma membrane, isolated from the supernatant of HSV-1-infected HOG cells were analyzed to find out whether they were involved in the viral cycle. The importance of our investigation lies in the detection, for the first time, of microvesicles containing HSV-1 virions. In addition, virus-containing microvesicles were endocytosed into CHO-K1 cells and were able to actively infect these otherwise nonpermissive cells. Finally, the infection of CHO cells with these virus-containing microvesicles was not completely neutralized by anti-HSV-1 antibodies, suggesting that these extracellular vesicles might shield the virus from neutralizing antibodies as a possible mechanism of immune evasion.


Asunto(s)
Micropartículas Derivadas de Células/virología , Herpes Simple/transmisión , Herpesvirus Humano 1/fisiología , Oligodendroglía/virología , Replicación Viral/fisiología , Animales , Anticuerpos Antivirales/inmunología , Células CHO , Línea Celular , Membrana Celular/metabolismo , Chlorocebus aethiops , Cricetulus , Endocitosis , Células HeLa , Herpes Simple/virología , Herpesvirus Humano 1/crecimiento & desarrollo , Humanos , Microscopía Electrónica de Transmisión , Oligodendroglía/citología , Células Vero , Internalización del Virus
8.
Cell Mol Life Sci ; 73(20): 3897-916, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27117550

RESUMEN

Alphavirus budding is driven by interactions between nucleocapsids assembled in the cytoplasm and envelope proteins present at the plasma membrane. So far, the expression of capsid and envelope proteins in infected cells has been considered an absolute requirement for alphavirus budding and propagation. In the present study, we show that Semliki Forest virus and Sindbis virus lacking the capsid gene can propagate in mammalian and insect cells. This propagation is mediated by the release of infectious microvesicles (iMVs), which are pleomorphic and have a larger size and density than wild-type virus. iMVs, which contain viral RNA inside and viral envelope proteins on their surface, are released at the plasma membrane and infect cells using the endocytic pathway in a similar way to wild-type virus. iMVs are not pathogenic in immunocompetent mice when injected intravenously, but can infect different organs like lungs and heart. Finally, we also show that alphavirus genomes without capsid can mediate the propagation of heterologous genes, making these vectors potentially interesting for gene therapy or vaccination studies. The minimalist infectious system described in this study shows that a self-replicating RNA able to express membrane proteins with binding and fusion properties is able to propagate, providing some insights into virus evolution.


Asunto(s)
Alphavirus/patogenicidad , Cápside/metabolismo , Membrana Celular/virología , Micropartículas Derivadas de Células/virología , Alphavirus/genética , Animales , Fusión Celular , Línea Celular , Micropartículas Derivadas de Células/metabolismo , Micropartículas Derivadas de Células/ultraestructura , Femenino , Genoma Viral , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones Endogámicos C57BL , Pruebas de Neutralización , ARN Viral/metabolismo , Virus de los Bosques Semliki/patogenicidad , Transfección , Proteínas del Envoltorio Viral/metabolismo , Proteínas Virales/metabolismo
9.
PLoS Pathog ; 10(4): e1004045, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24722773

RESUMEN

Coxsackievirus B3 (CVB3), a member of the picornavirus family and enterovirus genus, causes viral myocarditis, aseptic meningitis, and pancreatitis in humans. We genetically engineered a unique molecular marker, "fluorescent timer" protein, within our infectious CVB3 clone and isolated a high-titer recombinant viral stock (Timer-CVB3) following transfection in HeLa cells. "Fluorescent timer" protein undergoes slow conversion of fluorescence from green to red over time, and Timer-CVB3 can be utilized to track virus infection and dissemination in real time. Upon infection with Timer-CVB3, HeLa cells, neural progenitor and stem cells (NPSCs), and C2C12 myoblast cells slowly changed fluorescence from green to red over 72 hours as determined by fluorescence microscopy or flow cytometric analysis. The conversion of "fluorescent timer" protein in HeLa cells infected with Timer-CVB3 could be interrupted by fixation, suggesting that the fluorophore was stabilized by formaldehyde cross-linking reactions. Induction of a type I interferon response or ribavirin treatment reduced the progression of cell-to-cell virus spread in HeLa cells or NPSCs infected with Timer-CVB3. Time lapse photography of partially differentiated NPSCs infected with Timer-CVB3 revealed substantial intracellular membrane remodeling and the assembly of discrete virus replication organelles which changed fluorescence color in an asynchronous fashion within the cell. "Fluorescent timer" protein colocalized closely with viral 3A protein within virus replication organelles. Intriguingly, infection of partially differentiated NPSCs or C2C12 myoblast cells induced the release of abundant extracellular microvesicles (EMVs) containing matured "fluorescent timer" protein and infectious virus representing a novel route of virus dissemination. CVB3 virions were readily observed within purified EMVs by transmission electron microscopy, and infectious virus was identified within low-density isopycnic iodixanol gradient fractions consistent with membrane association. The preferential detection of the lipidated form of LC3 protein (LC3 II) in released EMVs harboring infectious virus suggests that the autophagy pathway plays a crucial role in microvesicle shedding and virus release, similar to a process previously described as autophagosome-mediated exit without lysis (AWOL) observed during poliovirus replication. Through the use of this novel recombinant virus which provides more dynamic information from static fluorescent images, we hope to gain a better understanding of CVB3 tropism, intracellular membrane reorganization, and virus-associated microvesicle dissemination within the host.


Asunto(s)
Micropartículas Derivadas de Células/virología , Enterovirus Humano B/fisiología , Infecciones por Enterovirus/metabolismo , Fagosomas/virología , Esparcimiento de Virus/fisiología , Animales , Micropartículas Derivadas de Células/genética , Micropartículas Derivadas de Células/metabolismo , Infecciones por Enterovirus/genética , Células HeLa , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Fagosomas/genética , Fagosomas/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
10.
Proteomics ; 13(10-11): 1608-23, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23505015

RESUMEN

Exosomes and microvesicles (MVs) are nanometer-sized, membranous vesicles secreted from many cell types into their surrounding extracellular space and into body fluids. These two classes of extracellular vesicles are regarded as a novel mechanism through which cancer cells, including virally infected cancer cells, regulate their micro-environment via the horizontal transfer of bioactive molecules: proteins, lipids, and nucleic acids (DNA, mRNA, micro-RNAs; oncogenic cargo hence often referred to as oncosomes). In head and neck cancer (HNC), exosomes and MVs have been described in Epstein Barr Virus (EBV)-associated nasopharyngeal cancer (NPC), as well as being positively correlated with oral squamous cell carcinoma (OSCC) progression. It has therefore been suggested that HNC-derived vesicles could represent a useful source for biomarker discovery, enriched in tumor antigens and cargo; hence fundamentally important for cancer progression. This current review offers an overall perspective on the roles of exosomes and MVs in HNC biology, focusing on EBV-associated NPC and OSCC. We also highlight the importance of saliva as a proximal and easily accessible bio-fluid for HNC detection, and propose that salivary vesicles might serve as an alternative model in the discovery of novel HNC biomarkers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Micropartículas Derivadas de Células/metabolismo , Exosomas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Animales , Micropartículas Derivadas de Células/virología , Exosomas/virología , Neoplasias de Cabeza y Cuello/virología , Herpesvirus Humano 4/genética , Humanos , MicroARNs/metabolismo , Proteoma/aislamiento & purificación , Proteoma/metabolismo , ARN Viral/metabolismo , Saliva/metabolismo
11.
J Clin Invest ; 122(12): 4685-97, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23160198

RESUMEN

Acute HIV-1 infection results in dysregulated immunity, which contributes to poor control of viral infection. DCs are key regulators of both adaptive and innate immune responses needed for controlling HIV-1, and we surmised that factors elicited during acute HIV-1 infection might impede DC function. We derived immature DCs from healthy donor peripheral blood monocytes and treated them with plasma from uninfected control donors and donors with acute HIV-1 infections. We found that the plasma from patients with HIV specifically inhibited DC function. This suppression was mediated by elevated apoptotic microparticles derived from dying cells during acute HIV-1 infection. Apoptotic microparticles bound to and inhibited DCs through the hyaluronate receptor CD44. These data suggest that targeting this CD44-mediated inhibition by apoptotic microparticles could be a novel strategy to potentiate DC activation of HIV-specific immunity.


Asunto(s)
Apoptosis , Micropartículas Derivadas de Células/inmunología , Células Dendríticas/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Receptores de Hialuranos/metabolismo , Micropartículas Derivadas de Células/virología , Células Dendríticas/fisiología , Células Dendríticas/virología , Infecciones por VIH/sangre , VIH-1/fisiología , Humanos , Receptores de Hialuranos/fisiología , Inmunidad Innata , Receptores Toll-Like/metabolismo , Viremia/virología
12.
J Immunol ; 189(2): 744-54, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22711894

RESUMEN

Exosomes and microvesicles (MV) are cell membranous sacs originating from multivesicular bodies and plasma membranes that facilitate long-distance intercellular communications. Their functional biology, however, remains incompletely understood. Macrophage exosomes and MV isolated by immunoaffinity and sucrose cushion centrifugation were characterized by morphologic, biochemical, and molecular assays. Lipidomic, proteomic, and cell biologic approaches uncovered novel processes by which exosomes and MV facilitate HIV-1 infection and dissemination. HIV-1 was "entrapped" in exosome aggregates. Robust HIV-1 replication followed infection with exosome-enhanced fractions isolated from infected cell supernatants. MV- and exosome-facilitated viral infections are affected by a range of cell surface receptors and adhesion proteins. HIV-1 containing exosomes readily completed its life cycle in human monocyte-derived macrophages but not in CD4(-) cells. The data support a significant role for exosomes as facilitators of viral infection.


Asunto(s)
Micropartículas Derivadas de Células/inmunología , Vesículas Citoplasmáticas/inmunología , Exosomas/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/patología , VIH-1/inmunología , Macrófagos/inmunología , Macrófagos/virología , Inmunidad Adaptativa , Animales , Micropartículas Derivadas de Células/química , Micropartículas Derivadas de Células/virología , Vesículas Citoplasmáticas/química , Vesículas Citoplasmáticas/virología , Exosomas/química , Exosomas/virología , Infecciones por VIH/etiología , VIH-1/patogenicidad , Células HeLa , Humanos , Inmunidad Innata , Macrófagos/patología , Ratones , Monocitos/inmunología , Monocitos/patología , Monocitos/virología
13.
J Acquir Immune Defic Syndr ; 59(4): 340-6, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22156911

RESUMEN

OBJECTIVE: HIV-1-infected patients have an increased risk for atherothrombosis and cardiovascular disease, but the mechanism behind these risks is poorly understood. We have previously reported that expression of tissue factor (TF) on circulating monocytes is increased in persons with HIV infection and that TF expression is related to immune activation, to levels of HIV in plasma, and to indices of microbial translocation. In this study, we explore the activation state of platelets in HIV disease. METHODS: Here, using flow cytometry-based assays, we measured platelet and platelet microparticle (PMP) activation in samples from HIV-1-infected donors and controls. RESULTS: Platelets and PMPs from HIV-1-infected patients are activated (as reflected by expression of CD62 P-selectin) and also more frequently expressed the procoagulant TF than did platelets and PMPs obtained from controls. Expression of these proteins was directly related to expression of TF on monocytes, to markers of T-cell activation (CD38 and HLA-DR), and to plasma levels of soluble CD14, the coreceptor for bacterial lipopolysaccharride. Platelet and microparticle expression of TF was not related to plasma levels of HIV but expression of P-selectin was related to plasma levels of HIV; neither TF nor P-selectin expression was related to CD4 T-cell count. CONCLUSIONS: Platelets and microparticles are activated in HIV infection, and this activated phenotype may contribute to the increased risk for cardiovascular and thrombotic events in this population although a role for other confounding cardiovascular risks cannot be completely excluded.


Asunto(s)
Plaquetas/virología , Micropartículas Derivadas de Células/virología , Infecciones por VIH/metabolismo , Selectina-P/biosíntesis , Activación Plaquetaria , Tromboplastina/biosíntesis , ADP-Ribosil Ciclasa 1/biosíntesis , Adulto , Anciano , Recuento de Linfocito CD4 , Femenino , Citometría de Flujo , Antígenos HLA-DR/biosíntesis , Humanos , Receptores de Lipopolisacáridos/sangre , Activación de Linfocitos , Masculino , Glicoproteínas de Membrana/biosíntesis , Persona de Mediana Edad , Monocitos/metabolismo , Regulación hacia Arriba , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...