Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 94(7)2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-31915281

RESUMEN

Human cytomegalovirus (HCMV) is a ubiquitous pathogen that encodes many proteins to modulate the host immune response. Extensive efforts have led to the elucidation of multiple strategies employed by HCMV to effectively block NK cell targeting of virus-infected cells and the major histocompatibility complex (MHC) class I-primed CD8+ T cell response. However, viral regulation of the MHC class II-mediated CD4+ T cell response is understudied in endogenous MHC class II-expressing cells, largely because the popular cell culture systems utilized for studying HCMV do not endogenously express MHC class II. Of the many cell types infected by HCMV in the host, myeloid cells, such as monocytes, are of particular importance due to their role in latency and subsequent dissemination throughout the host. We investigated the impact of HCMV infection on MHC class II in Kasumi-3 cells, a myeloid-progenitor cell line that endogenously expresses the MHC class II gene, HLA-DR. We observed a significant reduction in the expression of surface and total HLA-DR at 72 h postinfection (hpi) and 120 hpi in infected cells. The decrease in HLA-DR expression was independent of the expression of previously described viral genes that regulate the MHC class II complex or the unique short (US) region of HCMV, a region expressing many immunomodulatory genes. The altered surface level of HLA-DR was not a result of increased endocytosis and degradation but was a result of a reduction in HLA-DR transcripts due to a decrease in the expression of the class II transactivator (CIITA).IMPORTANCE Human cytomegalovirus (HCMV) is an opportunistic herpesvirus that is asymptomatic for healthy individuals but that can lead to severe pathology in patients with congenital infections and immunosuppressed patients. Thus, it is important to understand the modulation of the immune response by HCMV, which is understudied in the context of endogenous MHC class II regulation. Using Kasumi-3 cells as a myeloid progenitor cell model endogenously expressing MHC class II (HLA-DR), this study shows that HCMV decreases the expression of HLA-DR in infected cells by reducing the transcription of HLA-DR transcripts early during infection independently of the expression of previously implicated genes. This is an important finding, as it highlights a mechanism of immune evasion utilized by HCMV to decrease the expression of MHC class II in a relevant cell system that endogenously expresses the MHC class II complex.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Antígenos HLA-DR/genética , Células Progenitoras Mieloides/inmunología , Proteínas Nucleares/genética , Transactivadores/genética , Activación Transcripcional , Presentación de Antígeno , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Línea Celular , Citomegalovirus , Endocitosis , Fibroblastos/inmunología , Fibroblastos/virología , Regulación de la Expresión Génica , Humanos , Células Progenitoras Mieloides/virología
2.
Transfusion ; 59(9): 2938-2951, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31251408

RESUMEN

BACKGROUND: Dengue virus (DENV) is a significant threat to public health in tropical and subtropical regions, where the frequency of human migration is increasing. Transmission of DENV from donors to recipients after hematopoietic stem cell transplantation has been steadily described. However, the underlying mechanisms remain unclear. STUDY DESIGN AND METHODS: Freshly isolated bone marrow (BM) was subjected to DENV infection, followed by multicolor fluorescence-activated cell sorting (FACS) analysis. Virus in supernatants was collected and analyzed by plaque assay. RESULTS: DENV-1 to DENV-4 could effectively infect freshly obtained BM and produced infectious virus. DENV infection did not change the quantitative population of hematopoietic stem and progenitor cells (HSPCs), megakaryocytic progenitor cells (MkPs) and megakaryocytes. Additionally, DENV antigen, nonstructural protein 1, was enriched in HSPCs and MkPs of DENV infected marrow cells. CD34+, CD133+, or CD61+ cells sorted out from BM were not only the major contributing targets facilitating the DENV infection directly but also facilitated the spread of DENV into other cells when cocultured. CONCLUSION: Results suggest that DENV can efficiently infect HSPCs, which might jeopardize the recipients if DENV-infected cells were subsequently used. We therefore raise the need for DENV screening for both the donors and recipients of hematopoietic stem cell transplantation, especially for donors exposed to endemic areas, to mitigate DENV infection in immunocompromised recipients.


Asunto(s)
Virus del Dengue/crecimiento & desarrollo , Dengue/patología , Dengue/transmisión , Células Madre Hematopoyéticas/virología , Ensayo de Placa Viral , Antígenos Virales/análisis , Antígenos Virales/aislamiento & purificación , Células de la Médula Ósea/patología , Células de la Médula Ósea/fisiología , Células de la Médula Ósea/virología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Dengue/sangre , Virus del Dengue/patogenicidad , Sangre Fetal/citología , Sangre Fetal/virología , Citometría de Flujo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/fisiología , Humanos , Inmunofenotipificación , Megacariocitos/patología , Megacariocitos/fisiología , Megacariocitos/virología , Células Progenitoras Mieloides/patología , Células Progenitoras Mieloides/fisiología , Células Progenitoras Mieloides/virología
3.
J Virol ; 88(24): 13947-55, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25253336

RESUMEN

UNLABELLED: The UL111A gene of human cytomegalovirus encodes a viral homologue of the cellular immunomodulatory cytokine interleukin 10 (cIL-10), which, due to alternative splicing, results in expression of two isoforms designated LAcmvIL-10 (expressed during both lytic and latent infection) and cmvIL-10 (identified only during lytic infection). We have analyzed the functions of LAcmvIL-10 during latent infection of primary myeloid progenitor cells and found that LAcmvIL-10 is responsible, at least in part, for the known increase in secretion of cellular IL-10 and CCL8 in the secretomes of latently infected cells. This latency-associated increase in CCL8 expression results from a concomitant LAcmvIL-10-mediated suppression of the expression of the cellular microRNA (miRNA) hsa-miR-92a, which targets CCL8 directly. Taking the data together, we show that the previously observed downregulation of hsa-miR-92a and upregulation of CCL8 during HCMV latent infection of myeloid cells are intimately linked via the latency-associated expression of LAcmvIL-10. IMPORTANCE: HCMV latency causes significant morbidity and mortality in immunocompromised individuals, yet HCMV is carried silently (latently) in 50 to 90% of the population. Understanding how HCMV maintains infection for the lifetime of an infected individual is critical for the treatment of immunocompromised individuals suffering with disease as a result of HCMV. In this study, we analyze one of the proteins that are expressed during the "latent" phase of HCMV, LAcmvIL-10, and find that the expression of the gene modulates the microenvironment of the infected cell, leading to evasion of the immune system.


Asunto(s)
Quimiocina CCL8/metabolismo , Citomegalovirus/fisiología , Interleucina-10/metabolismo , MicroARNs/metabolismo , Proteínas Virales/metabolismo , Latencia del Virus , Células Cultivadas , Humanos , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/virología
4.
J Virol ; 86(18): 9854-65, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22761372

RESUMEN

Human cytomegalovirus (HCMV) is a herpesvirus that establishes a lifelong, latent infection within a host. At times when the immune system is compromised, the virus undergoes a lytic reactivation producing infectious progeny. The identification and understanding of the biological mechanisms underlying HCMV latency and reactivation are not completely defined. To this end, we have developed a tractable in vitro model system to investigate these phases of viral infection using a clonal population of myeloid progenitor cells (Kasumi-3 cells). Infection of these cells results in maintenance of the viral genome with restricted viral RNA expression that is reversed with the addition of the phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA, also known as PMA). Additionally, a latent viral transcript (LUNA) is expressed at times where viral lytic transcription is suppressed. Infected Kasumi-3 cells initiate production of infectious virus following TPA treatment, which requires cell-to-cell contact for efficient transfer of virus to other cell types. Importantly, lytically infected fibroblast, endothelial, or epithelial cells can transfer virus to Kasumi-3 cells, which fail to initiate lytic replication until stimulated with TPA. Finally, inflammatory cytokines, in addition to the pharmacological agent TPA, are sufficient for transcription of immediate-early (IE) genes following latent infection. Taken together, our findings argue that the Kasumi-3 cell line is a tractable in vitro model system with which to study HCMV latency and reactivation.


Asunto(s)
Citomegalovirus/fisiología , Citomegalovirus/patogenicidad , Células Progenitoras Mieloides/virología , Secuencia de Bases , Línea Celular , Citocinas/fisiología , Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , ADN Viral/genética , Expresión Génica/efectos de los fármacos , Genes Inmediatos-Precoces , Genoma Viral , Humanos , Mediadores de Inflamación/fisiología , Modelos Biológicos , Células Progenitoras Mieloides/efectos de los fármacos , Células Progenitoras Mieloides/fisiología , Acetato de Tetradecanoilforbol/farmacología , Activación Viral/genética , Activación Viral/fisiología , Latencia del Virus/genética , Latencia del Virus/fisiología , Replicación Viral/genética , Replicación Viral/fisiología
5.
Allergy ; 68(5): 681-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23573902

RESUMEN

BACKGROUND: In drug-induced hypersensitivity syndrome (DIHS), latent human herpesvirus (HHV)-6 is frequently reactivated in association with flaring of symptoms such as fever and hepatitis. We recently demonstrated an emergence of monomyeloid precursors expressing HHV-6 antigen in the circulation during this clinical course. METHODS: To clarify the mechanism of HHV-6 reactivation, we immunologically investigated peripheral blood mononuclear cells (PBMCs), skin-infiltrating cells, and lymphocytes expanded from skin lesions of patients with DIHS. RESULTS: The circulating monomyeloid precursors in the patients with DIHS were mostly CD11b(+) CD13(+) CD14(-) CD16(high) and showed substantial expression of skin-associated molecules, such as CCR4. CD13(+) CD14(-) cells were also found in the DIHS skin lesions, suggesting skin recruitment of this cell population. We detected high levels of high-mobility group box (HMGB)-1 in blood and skin lesions in the active phase of patients with DIHS and showed that recombinant HMGB-1 had functional chemoattractant activity for monocytes/monomyeloid precursors in vitro. HHV-6 infection of the skin-resident CD4(+) T cells was confirmed by the presence of its genome and antigen. This infection was likely to be mediated by monomyeloid precursors recruited to the skin, because normal CD4(+) T cells gained HHV-6 antigen after in vitro coculture with highly virus-loaded monomyeloid precursors from the patients. CONCLUSIONS: Our results suggest that monomyeloid precursors harboring HHV-6 are navigated by HMGB-1 released from damaged skin and probably cause HHV-6 transmission to skin-infiltrating CD4(+) T cells, which is an indispensable event for HHV-6 replication. These findings implicate the skin as a cryptic and primary site for initiating HHV-6 reactivation.


Asunto(s)
Hipersensibilidad a las Drogas/etiología , Herpesvirus Humano 6/fisiología , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/virología , Piel/inmunología , Piel/patología , Activación Viral , Adulto , Anciano , Anciano de 80 o más Años , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Erupciones por Medicamentos , Femenino , Proteína HMGB1/sangre , Humanos , Masculino , Persona de Mediana Edad , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/ultraestructura , Fenotipo , Infecciones por Roseolovirus/transmisión
6.
J Immunol ; 186(7): 4314-24, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21357269

RESUMEN

We previously showed that the TLR7/8 agonist, R-848, activated HIV from cells of myeloid-monocytic origin. In this work, we show that this effect was solely due to triggering TLR8 and that NF-κB was involved in the TLR8-mediated activation of HIV from latently infected cells of myeloid-monocytic origin. Inhibition of Erk1/2 or p38α resulted in attenuation of TLR8-mediated activation of NF-κB. Western blots confirmed that TLR8 triggering activated Erk1/2 and p38α but, surprisingly, not JNK. Although the Erk1/2 inhibitors resulted in a less attenuated TLR8-mediated NF-κB response than did p38α inhibitors, they had a more pronounced effect on blocking TLR8-mediated HIV replication, indicating that other transcription factors controlled by Erk1/2 are involved in TLR8-mediated HIV activation from latently infected cells. TNF-α, which was secreted subsequent to TLR8 triggering, contributed to the activation of HIV from the latently infected cells in an autocrine manner, revealing a bimodal mechanism by which the effect of TLR8 triggering can be sustained. We also found that TNF-α secreted by myeloid dendritic cells acted in a paracrine manner in the activation of HIV from neighboring latently infected CD4(+) T cells, which do not express TLR8. Notably, monocytes from highly active antiretroviral therapy-treated HIV(+) patients with suppressed HIV RNA showed a robust TNF-α secretion in response to TLR8 agonists, pointing to a functional TLR8 signaling axis in HIV infection. Thus, triggering TLR8 represents a very promising strategy for attacking the silent HIV from its reservoir in HIV(+) patients treated successfully with highly active antiretroviral therapy.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , VIH-1/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Monocitos/inmunología , Monocitos/virología , Receptor Toll-Like 8/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Activación Viral/inmunología , Linfocitos T CD4-Positivos/enzimología , Línea Celular , Células Cultivadas , Células Clonales , Células HEK293 , Humanos , Monocitos/enzimología , Células Progenitoras Mieloides/enzimología , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/virología , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 7/fisiología , Receptor Toll-Like 8/agonistas
7.
Blood ; 114(19): 4128-37, 2009 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-19706889

RESUMEN

The capacity of human cytomegalovirus (HCMV) to establish and maintain a latent infection from which it can later reactivate ensures its widespread distribution in the population, but the mechanisms enabling maintenance of latency in the face of a robust immune system are poorly understood. We examined the role of the HCMV UL111A gene, which encodes homologs of the immunosuppressive cytokine interleukin-10 in the context of latent infection of myeloid progenitor cells. A UL111A deletion virus was able to establish, maintain, and reactivate from experimental latency in a manner comparable with parental virus, but major histocompatibility complex class II levels increased significantly on the surfaces of cells infected with the deletion virus. Importantly, there was an increase in both allogeneic and autologous peripheral blood mononuclear cells and CD4(+) T-cell responses to UL111A deletion virus-infected myeloid progenitors, indicating that loss of the capacity to express viral interleukin-10 during latency results in latently infected cells becoming more readily recognizable by a critical arm of the immune response. The detection of a viral gene that suppresses CD4(+) T-cell recognition of latently infected cells identifies an immune evasion strategy that probably enhances the capacity of HCMV to persist in a latent state within the human host.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Citomegalovirus/genética , Citomegalovirus/inmunología , Genes Virales , Latencia del Virus/inmunología , Autoantígenos , Citomegalovirus/patogenicidad , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Regulación hacia Abajo , Eliminación de Gen , Antígenos de Histocompatibilidad Clase II/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Técnicas In Vitro , Isoantígenos , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/virología
8.
Virol J ; 8: 397, 2011 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-21831263

RESUMEN

BACKGROUND: Human immunodeficiency virus type 1 (HIV-1) viral protein R (Vpr) is a virion-associated regulatory protein that functions at several points within the viral life cycle and has been shown to accumulate primarily in the nucleus and at the nuclear envelope. However, most studies have investigated Vpr localization employing cell types irrelevant to HIV-1 pathogenesis. To gain a better understanding of how cellular phenotype might impact HIV-1 Vpr intracellular localization, Vpr localization was examined in several cell lines representing major cellular targets for HIV-1 infection within the peripheral blood, bone marrow, and central nervous system (CNS). RESULTS: Utilizing a green fluorescent protein-tagged Vpr, we detected Vpr mainly in foci inside the nucleus, at the nuclear envelope, and around the nucleoli, with dispersed accumulation in the cytoplasm of human endothelial kidney 293T cells. No differences were observed in Vpr localization pattern with respect to either the location of the tag (N- or C-terminus) or the presence of other viral proteins. Subsequently, the Vpr localization pattern was explored in two primary HIV-1 target cells within the peripheral blood: the CD4+ T lymphocyte (represented by the Jurkat CD4+ T-cell line) and the monocyte-macrophage (represented by the U-937 cell line). Vpr was found primarily in speckles within the cytoplasm of the Jurkat T cells, whereas it accumulated predominantly intranuclearly in U-937 monocytic cells. These patterns differ from that observed in a bone marrow progenitor cell line (TF-1), wherein Vpr localized mainly at the nuclear envelope with some intranuclear punctuate staining. Within the CNS, we examined two astroglioma cell lines and found that Vpr displayed a perinuclear and cytoplasmic distribution. CONCLUSIONS: The results suggest that the pattern of Vpr localization depends on cellular phenotype, probably owing to interactions between Vpr and cell type-specific host factors. These interactions, in turn, are likely coupled to specific roles that Vpr plays in each cell type within the context of the viral life cycle. Phenotype-specific Vpr localization patterns might also provide an explanation with respect to Vpr secretion or release from HIV-1-infected cells within the peripheral blood and CNS.


Asunto(s)
Productos del Gen vpr/análisis , VIH-1/patogenicidad , Interacciones Huésped-Patógeno , Astrocitos/química , Astrocitos/virología , Linfocitos T CD4-Positivos/química , Linfocitos T CD4-Positivos/virología , Línea Celular , Nucléolo Celular/química , Núcleo Celular/química , Citoplasma/química , Células Endoteliales/química , Células Endoteliales/virología , Humanos , Macrófagos/química , Macrófagos/virología , Células Progenitoras Mieloides/química , Células Progenitoras Mieloides/virología , Membrana Nuclear/química
9.
J Immunol ; 181(6): 4010-8, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18768856

RESUMEN

Dendritic cells (DCs) are a diverse population with the capacity to respond to a variety of pathogens. Because of their critical role in pathogenesis and Ag-specific adaptive immune responses, DCs are the focus of extensive study and incorporation into a variety of immunotherapeutic strategies. The diversity of DC subsets imposes a substantial challenge to the successful development of DC-based therapies, requiring identification of the involved subset(s) and the potential roles each contributes to the immunologic responses. The recently developed and promising Venezuelan equine encephalitis replicon particle (VRP) vector system has conserved tropism for a subset of myeloid DCs. This immunotherapeutic vector permits in situ targeting of DCs; however, it targets a restricted subset of DCs, which are heretofore uncharacterized. Using a novel technique, we isolated VRP-receptive and -nonreceptive populations from human monocyte-derived DCs. Comparative gene expression analysis revealed significant differential gene expression, supporting the existence of two distinct DC populations. Further analysis identified constitutive expression of the proinflammatory cytokine IL-32 as a distinguishing characteristic of VRP-receptive DCs. IL-32 transcript was exclusively expressed (>50 fold) in the VRP-receptive DC population relative to the background level of expression in the nonreceptive population. The presence of IL-32 transcript was accompanied by protein expression. These data are the first to identify a subset of immature monocyte-derived DCs constitutively expressing IL-32 and they provide insights into both DC biology and potential mechanisms employed by this potent vector system.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/virología , Virus de la Encefalitis Equina Venezolana/inmunología , Interleucinas/biosíntesis , Interleucinas/genética , Replicón/inmunología , Sitios de Unión/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Membrana Celular/inmunología , Membrana Celular/metabolismo , Membrana Celular/virología , Separación Celular , Células Dendríticas/citología , Virus de la Encefalitis Equina Venezolana/patogenicidad , Perfilación de la Expresión Génica , Humanos , Interleucinas/fisiología , Monocitos/citología , Monocitos/inmunología , Monocitos/virología , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/virología , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/biosíntesis , Replicón/genética , Transcripción Genética , Transducción Genética
10.
DNA Cell Biol ; 26(6): 387-94, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17570762

RESUMEN

Cells of the monocyte-macrophage lineage play an important role in human immunodeficiency virus type 1 (HIV-1)-associated disease. Infected myeloid precursor cells of the bone marrow are thought to be a viral reservoir that may repopulate the peripheral blood, central nervous system (CNS), and other organ systems throughout the course of disease. To model select aspects of HIV-1 infection of the bone marrow compartment in vitro, the erythro-myeloid precursor cell line, TF-1, was used. Phorbol 12-myristate 13-acetate (PMA) was found to induce the TF-1 cell line to differentiate through the myeloid lineage and become activated, as demonstrated by cellular morphologic changes and surface expression of differentiation and activation markers. Herein we demonstrate that HIV-1 long terminal repeats (LTRs) from T-, M-, and dual-tropic molecular clones have similar basal LTR activity in TF-1 cells and that differentiation of these cells by PMA resulted in increased LTR activity. Examination of specific cis-acting elements involved in basal and PMA-induced LTR activity demonstrated that the transcription factor families nuclear factor-kappa B (NF-kappaB) and specificity protein (Sp) contributed to the LTR activity of TF-1 cells, the Sp family being the most critical. These studies elucidate the impact of infected bone marrow monocytic cell differentiation on LTR activity and its potential impact on HIV-1-associated disease.


Asunto(s)
Duplicado del Terminal Largo de VIH , VIH-1/genética , Células Progenitoras Mieloides/virología , Antígenos CD/metabolismo , Secuencia de Bases , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , ADN Viral/genética , Humanos , Técnicas In Vitro , Mutagénesis Sitio-Dirigida , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/efectos de los fármacos , Células Progenitoras Mieloides/metabolismo , FN-kappa B/metabolismo , Factores de Transcripción Sp/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Transcripción Genética/efectos de los fármacos
11.
Methods Mol Biol ; 334: 199-209, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16861765

RESUMEN

Cytomegalovirus latency depends on an interaction with hematopoietic cells in bone marrow and peripheral blood. The distribution of latent viral DNA and transcripts in these cells was investigated using methods based on polymerase chain reaction (PCR)-driven in situ hybridization (ISH) and reverse transcription (RT)-PCR-driven ISH. Using a conventional thermal cycler, latent viral DNA or transcripts were amplified within suspension cells. Amplified products were then detected by nonisotopic ISH on cells cytospun on glass microscope slides. During experimental latent infection of cultured granulocyte-macrophage progenitors, the viral genome was detected in more than 90% of cells. During natural infection, viral genomes were detected in 0.004 to 0.01% of mono-nuclear cells from granulocyte colony-stimulating factor mobilized peripheral blood or bone marrow from healthy seropositive donors. When evaluated by RT-PCR-ISH, only a small proportion of experimentally infected cells (approx 2%) had detectable latent transcripts. The application of PCR-ISH and RT-PCR-ISH has enabled the identification of the small percentage of bone marrow-derived mononuclear cells that become latently infected during natural infection and suggests that latency may proceed in some cells that fail to encode latent transcripts.


Asunto(s)
Citomegalovirus/genética , Citomegalovirus/aislamiento & purificación , ADN Viral/análisis , ADN Viral/genética , Células Progenitoras Mieloides/virología , Reacción en Cadena de la Polimerasa/métodos , Latencia del Virus/genética , Citomegalovirus/crecimiento & desarrollo , Humanos , Hibridación in Situ , ARN Viral/análisis , ARN Viral/genética
12.
Res Vet Sci ; 78(2): 151-4, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15563922

RESUMEN

Molecularly cloned feline leukemia virus (FeLV)-clone 33 (C-33), derived from a cat with acute myelocytic leukemia (AML), was examined to assess its relation to the pathogenesis of AML and myelodysplastic syndrome (MDS). To evaluate in vitro pathogenicity of FeLV C-33, bone marrow colony-forming assay was performed on marrow cells infected with FeLV C-33 or an FeLV subgroup A strain (61E, a molecularly cloned strain with minimal pathogenicity). The myeloid colony-forming activity of feline bone marrow mononuclear cells infected with FeLV C-33 was significantly lower than that of cells infected with 61E. This suggests that FeLV C-33 has myeloid lineage-specific pathogenicity for cats, and that FeLV C-33 infection is useful as an experimental model for investigating pathogenesis of MDS and AML.


Asunto(s)
Enfermedades de los Gatos/virología , Virus de la Leucemia Felina/genética , Virus de la Leucemia Felina/patogenicidad , Leucemia Mieloide Aguda/veterinaria , Células Progenitoras Mieloides/virología , Infecciones por Retroviridae/veterinaria , Secuencias Repetidas Terminales , Infecciones Tumorales por Virus/veterinaria , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/virología , Gatos , Clonación Molecular , ADN Viral/química , ADN Viral/genética , Leucemia Mieloide Aguda/patología , Leucemia Mieloide Aguda/virología , Síndromes Mielodisplásicos/veterinaria , Síndromes Mielodisplásicos/virología , Células Progenitoras Mieloides/citología , Reacción en Cadena de la Polimerasa/veterinaria , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología
13.
Virology ; 446(1-2): 173-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24074579

RESUMEN

Erythrovirus B19 (B19V) is a member of the family Parvoviridae. Infection with B19V has been linked to a variety of diseases including erythroid, thyroid, neurological and autoimmune diseases. Here we show that infection of primary CD36+ cells with B19V coincides with downregulation of thyroid, retinoid, and estrogen hormone receptors. In addition we show changes in expression of a variety of related downstream signaling genes participating in cancer and cardiac-related diseases in B19V-infected erythroid primary cells.


Asunto(s)
Interacciones Huésped-Patógeno , Células Progenitoras Mieloides/virología , Parvovirus B19 Humano/fisiología , Receptores de Estrógenos/biosíntesis , Receptores de Ácido Retinoico/biosíntesis , Receptores de Hormona Tiroidea/biosíntesis , Replicación Viral , Antígenos CD36/análisis , Células Cultivadas , Regulación hacia Abajo , Humanos , Células Progenitoras Mieloides/química , Transducción de Señal
15.
Indian J Med Microbiol ; 26(3): 252-5, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18695326

RESUMEN

This study examined the incidence of human herpes virus-6 (HHV-6) and human cytomegalovirus (HCMV) infections that are potentially transmitted to haematopoietic stem cells (HSC) transplant recipients via bone marrow (BM) or umbilical cord blood (UCB). Bone marrow progenitor cells were collected from 30 allogenic BM donors. UCB HSC were collected from 34 subjects. The extracted DNA was then processed using nested polymerase chain reaction (nPCR) technique. HCMV and HHV-6 serological status were determined by enzyme immunoassay (EIA). Nested PCR identified HCMV in 22 (73%) of 30 samples of BM progenitor cells but in only eight (23.5%) of 34 samples of UBC HSC ( P = 0.001). HHV-6 DNA was detected in 11 (36.6%) of 30 BM progenitor cells and in only one (2.9%) of 34 UBC cells ( P = 0.002). Both HHV-6 and HCMV infections were determined in nine (26.5%) of 34 bone marrow samples. The results indicate that, the risk of HCMV and HHV-6 via BM progenitor cells is higher than transmission by UCB cells ( P= 0.04).


Asunto(s)
Médula Ósea/virología , Citomegalovirus/aislamiento & purificación , Sangre Fetal/virología , Células Madre Hematopoyéticas/virología , Herpesvirus Humano 6/aislamiento & purificación , Células Progenitoras Mieloides/virología , Adulto , Anticuerpos Antivirales/sangre , Citomegalovirus/genética , Infecciones por Citomegalovirus , ADN Viral/genética , Femenino , Herpesvirus Humano 6/genética , Humanos , Técnicas para Inmunoenzimas , Incidencia , Masculino , Reacción en Cadena de la Polimerasa , Infecciones por Roseolovirus/epidemiología
16.
J Virol ; 81(13): 7274-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17428873

RESUMEN

Infection with a recombinant murine-feline gammaretrovirus, MoFe2, or with the parent virus, Moloney murine leukemia virus, caused significant reduction in B-lymphoid differentiation of bone marrow at 2 to 8 weeks postinfection. The suppression was selective, in that myeloid potential was significantly increased by infection. Analysis of cell surface markers and immunoglobulin H gene rearrangements in an in vitro model demonstrated normal B-lymphoid differentiation after infection but significantly reduced viability of differentiating cells. This reduction in viability may confer a selective advantage on undifferentiated lymphoid progenitors in the bone marrow of gammaretrovirus-infected animals and thereby contribute to the establishment of a premalignant state.


Asunto(s)
Linfocitos B/metabolismo , Diferenciación Celular , Virus de la Leucemia Felina/metabolismo , Virus de la Leucemia Murina de Moloney/metabolismo , Células Progenitoras Mieloides/metabolismo , Infecciones por Retroviridae/metabolismo , Infecciones Tumorales por Virus/mortalidad , Animales , Linfocitos B/patología , Linfocitos B/virología , Médula Ósea/metabolismo , Médula Ósea/patología , Médula Ósea/virología , Gatos , Hematopoyesis Extramedular , Ratones , Células Progenitoras Mieloides/patología , Células Progenitoras Mieloides/virología , Infecciones por Retroviridae/patología , Infecciones Tumorales por Virus/patología
17.
Blood ; 108(12): 3691-9, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16931631

RESUMEN

Human cytomegalovirus (HCMV) establishes and maintains a latent infection in myeloid cells and can reactivate to cause serious disease in allograft recipients. To better understand the molecular events associated with the establishment of latency, we tracked the virus following infection of primary human myeloid progenitor cells at days 1, 2, 3, 5, and 11. At all time points, the viral genome was maintained in most cells at approximately 10 copies. Infectious virus was not detected, but virus could be reactivated by extended fibroblast coculture. In contrast to wild-type HCMV, the viral genome was rapidly lost from myeloid progenitors infected with ultraviolet (UV)-inactivated virus, suggesting viral gene expression was required for efficient establishment of latency. To identify viral genes associated with the establishment phase, RNA from each time point was interrogated using custom-made HCMV gene microarrays. Using this approach, we detected expression of viral RNAs at all time points. The pattern of expression differed from that which occurs during productive infection, and decreased over time. This study provides evidence that a molecular pathway into latency is associated with expression of a unique subset of viral transcripts. Viral genes expressed during the establishment phase may serve as targets for therapies to interrupt this process.


Asunto(s)
Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/fisiología , Regulación Viral de la Expresión Génica/fisiología , Células Progenitoras Mieloides/virología , Activación Viral/fisiología , Latencia del Virus/fisiología , Células Cultivadas , Técnicas de Cocultivo , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/prevención & control , Fibroblastos/metabolismo , Fibroblastos/virología , Perfilación de la Expresión Génica/métodos , Regulación Viral de la Expresión Génica/efectos de la radiación , Genoma Viral/fisiología , Genoma Viral/efectos de la radiación , Humanos , Células Progenitoras Mieloides/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Viral/metabolismo , Trasplante de Células Madre/efectos adversos , Trasplante de Células Madre/métodos , Rayos Ultravioleta , Terapia Ultravioleta/métodos , Activación Viral/efectos de la radiación , Inactivación de Virus/efectos de la radiación , Latencia del Virus/efectos de la radiación
18.
Scand J Immunol ; 61(6): 545-50, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15963049

RESUMEN

Due to the complexity and redundancy of molecular processes governing the development and function of haematopoietic cells, experimental procedures allowing simultaneous alteration in gene expression of multiple genes in vivo are needed. Here, we describe a protocol allowing for simultaneous transduction of haematopoietic stem cells (HSC) with two different replication incompetent retroviral expression vectors followed by transplantation of lethally irradiated recipient mice. These bicistronic retroviral vectors carried genes for the enhanced green and yellow florescent proteins (EGFP and EYFP) respectively. Spleen cells from reconstituted animals were stained for common lymphocyte and myeloid markers, then analysed on a two-laser, 488 and 635 nm, flow cytometer equipped with a 510/20-nm bandpass filter for FL1, a 550/30-nm bandpass filter for FL2 and a 530-nm short-pass dichroic mirror. It was demonstrated that cells expressing EGFP, EYFP or combinations thereof could be distinguished and analysed for staining with PerCP- and APC-conjugated reagents. We found that a sizable proportion of cells (70%) from reconstituted animals expressed EGFP and/or EYFP and that expression of these genes did not affect lymphoid or myeloid development. We also demonstrated that the alternative optical configuration allowed for conventional multiparameter flow cytometric analyses.


Asunto(s)
Trasplante de Médula Ósea , Vectores Genéticos , Células Madre Hematopoyéticas/metabolismo , Linfocitos/metabolismo , Retroviridae/genética , Transducción Genética , Animales , Femenino , Citometría de Flujo , Expresión Génica , Células Madre Hematopoyéticas/virología , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Linfocitos/virología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/virología , Bazo/inmunología
19.
Zhonghua Xue Ye Xue Za Zhi ; 20(3): 143-5, 1999 Mar.
Artículo en Zh | MEDLINE | ID: mdl-11601242

RESUMEN

OBJECTIVE: To investigate whether the suppression of CFU-GM is caused directly by human cytomegalovirus(HCMV). METHODS: The effect of HCMV AD 169 strain on CFU-GM growth was assayed in vitro and HCMV AD 169 DNA in CFU-GM was detected by in situ PCR(ISPCR). RESULTS: Significant inhibition of CFU-GM was observed when HCMV AD 169 containing fluid was diluted to 2 x 10(5) pfu/ml and 2 x 10(6) pfu/ml (P < 0.01). The suppression was in a dose-dependent fashion. HCMV AD 169-DNA was detected by ISPCR on the nuclear of CFU-GM. CONCLUSION: HCMV might cause suppression of CFU-GM growth by direct infection.


Asunto(s)
Citomegalovirus/patogenicidad , Células Progenitoras Mieloides/citología , Adolescente , Adulto , División Celular , Femenino , Humanos , Masculino , Persona de Mediana Edad , Células Progenitoras Mieloides/virología
20.
J Virol ; 78(8): 4054-62, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15047822

RESUMEN

Herpesviruses establish lifelong latent infections in their hosts. Human cytomegalovirus (CMV) targets a population of bone marrow-derived myeloid lineage progenitor cells that serve as a reservoir for reactivation; however, the mechanisms by which latent CMV infection is maintained are unknown. To gain insights into mechanisms of maintenance and reactivation, we employed microarrays of approximately 26,900 sequence-verified human cDNAs to assess global changes in cellular gene expression during experimental CMV latent infection of granulocyte-macrophage progenitors (GM-Ps). This analysis revealed at least 29 host cell genes whose expression was increased and six whose expression was decreased during CMV latency. These changes in transcript levels appeared to be authentic, judging on the basis of further analysis of a subset by semiquantitative reverse transcription-PCR. This study provides a comprehensive snapshot of changes in host cell gene expression that result from latent infection and suggest that CMV regulates genes that encode proteins involved in immunity and host defense, cell growth, signaling, and transcriptional regulation. The host genes whose expression we found altered are likely to contribute to an environment that sustains latent infection.


Asunto(s)
Citomegalovirus/patogenicidad , Células Progenitoras Mieloides/fisiología , Células Progenitoras Mieloides/virología , Secuencia de Bases , Células Cultivadas , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , ADN Complementario/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA