Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Virol ; 94(7)2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-31915281

RESUMO

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.


Assuntos
Infecções por Citomegalovirus/imunologia , Antígenos HLA-DR/genética , Células Progenitoras Mieloides/imunologia , Proteínas Nucleares/genética , Transativadores/genética , Ativação Transcricional , Apresentação de Antígeno , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Linhagem Celular , Citomegalovirus , Endocitose , Fibroblastos/imunologia , Fibroblastos/virologia , Regulação da Expressão Gênica , Humanos , Células Progenitoras Mieloides/virologia
2.
Transfusion ; 59(9): 2938-2951, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31251408

RESUMO

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.


Assuntos
Vírus da Dengue/crescimento & desenvolvimento , Dengue/patologia , Dengue/transmissão , Células-Tronco Hematopoéticas/virologia , Ensaio de Placa Viral , Antígenos Virais/análise , Antígenos Virais/isolamento & purificação , Células da Medula Óssea/patologia , Células da Medula Óssea/fisiologia , Células da Medula Óssea/virologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Dengue/sangue , Vírus da Dengue/patogenicidade , Sangue Fetal/citologia , Sangue Fetal/virologia , Citometria de Fluxo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/patologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Imunofenotipagem , Megacariócitos/patologia , Megacariócitos/fisiologia , Megacariócitos/virologia , Células Progenitoras Mieloides/patologia , Células Progenitoras Mieloides/fisiologia , Células Progenitoras Mieloides/virologia
3.
J Virol ; 88(24): 13947-55, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25253336

RESUMO

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.


Assuntos
Quimiocina CCL8/metabolismo , Citomegalovirus/fisiologia , Interleucina-10/metabolismo , MicroRNAs/metabolismo , Proteínas Virais/metabolismo , Latência Viral , Células Cultivadas , Humanos , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/virologia
4.
Virology ; 446(1-2): 173-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24074579

RESUMO

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.


Assuntos
Interações Hospedeiro-Patógeno , Células Progenitoras Mieloides/virologia , Parvovirus B19 Humano/fisiologia , Receptores de Estrogênio/biossíntese , Receptores do Ácido Retinoico/biossíntese , Receptores dos Hormônios Tireóideos/biossíntese , Replicação Viral , Antígenos CD36/análise , Células Cultivadas , Regulação para Baixo , Humanos , Células Progenitoras Mieloides/química , Transdução de Sinais
5.
Allergy ; 68(5): 681-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23573902

RESUMO

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.


Assuntos
Hipersensibilidade a Drogas/etiologia , Herpesvirus Humano 6/fisiologia , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/virologia , Pele/imunologia , Pele/patologia , Ativação Viral , Adulto , Idoso , Idoso de 80 Anos ou mais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Toxidermias , Feminino , Proteína HMGB1/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/ultraestrutura , Fenótipo , Infecções por Roseolovirus/transmissão
6.
J Virol ; 86(18): 9854-65, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22761372

RESUMO

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.


Assuntos
Citomegalovirus/fisiologia , Citomegalovirus/patogenicidade , Células Progenitoras Mieloides/virologia , Sequência de Bases , Linhagem Celular , Citocinas/fisiologia , Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , DNA Viral/genética , Expressão Gênica/efeitos dos fármacos , Genes Precoces , Genoma Viral , Humanos , Mediadores da Inflamação/fisiologia , Modelos Biológicos , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/fisiologia , Acetato de Tetradecanoilforbol/farmacologia , Ativação Viral/genética , Ativação Viral/fisiologia , Latência Viral/genética , Latência Viral/fisiologia , Replicação Viral/genética , Replicação Viral/fisiologia
7.
Virol J ; 8: 397, 2011 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-21831263

RESUMO

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.


Assuntos
Produtos do Gene vpr/análise , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Astrócitos/química , Astrócitos/virologia , Linfócitos T CD4-Positivos/química , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Nucléolo Celular/química , Núcleo Celular/química , Citoplasma/química , Células Endoteliais/química , Células Endoteliais/virologia , Humanos , Macrófagos/química , Macrófagos/virologia , Células Progenitoras Mieloides/química , Células Progenitoras Mieloides/virologia , Membrana Nuclear/química
8.
J Immunol ; 186(7): 4314-24, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21357269

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , HIV-1/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Monócitos/imunologia , Monócitos/virologia , Receptor 8 Toll-Like/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Ativação Viral/imunologia , Linfócitos T CD4-Positivos/enzimologia , Linhagem Celular , Células Cultivadas , Células Clonais , Células HEK293 , Humanos , Monócitos/enzimologia , Células Progenitoras Mieloides/enzimologia , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/virologia , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/fisiologia , Receptor 8 Toll-Like/agonistas
9.
Blood ; 114(19): 4128-37, 2009 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-19706889

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Citomegalovirus/genética , Citomegalovirus/imunologia , Genes Virais , Latência Viral/imunologia , Autoantígenos , Citomegalovirus/patogenicidade , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Regulação para Baixo , Deleção de Genes , Antígenos de Histocompatibilidade Classe II/metabolismo , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Técnicas In Vitro , Isoantígenos , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/virologia
10.
J Immunol ; 181(6): 4010-8, 2008 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-18768856

RESUMO

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.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Vírus da Encefalite Equina Venezuelana/imunologia , Interleucinas/biossíntese , Interleucinas/genética , Replicon/imunologia , Sítios de Ligação/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Membrana Celular/imunologia , Membrana Celular/metabolismo , Membrana Celular/virologia , Separação Celular , Células Dendríticas/citologia , Vírus da Encefalite Equina Venezuelana/patogenicidade , Perfilação da Expressão Gênica , Humanos , Interleucinas/fisiologia , Monócitos/citologia , Monócitos/imunologia , Monócitos/virologia , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/virologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/biossíntese , Replicon/genética , Transcrição Gênica , Transdução Genética
11.
Indian J Med Microbiol ; 26(3): 252-5, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18695326

RESUMO

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).


Assuntos
Medula Óssea/virologia , Citomegalovirus/isolamento & purificação , Sangue Fetal/virologia , Células-Tronco Hematopoéticas/virologia , Herpesvirus Humano 6/isolamento & purificação , Células Progenitoras Mieloides/virologia , Adulto , Anticorpos Antivirais/sangue , Citomegalovirus/genética , Infecções por Citomegalovirus , DNA Viral/genética , Feminino , Herpesvirus Humano 6/genética , Humanos , Técnicas Imunoenzimáticas , Incidência , Masculino , Reação em Cadeia da Polimerase , Infecções por Roseolovirus/epidemiologia
12.
DNA Cell Biol ; 26(6): 387-94, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17570762

RESUMO

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.


Assuntos
Repetição Terminal Longa de HIV , HIV-1/genética , Células Progenitoras Mieloides/virologia , Antígenos CD/metabolismo , Sequência de Bases , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , DNA Viral/genética , Humanos , Técnicas In Vitro , Mutagênese Sítio-Dirigida , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/metabolismo , NF-kappa B/metabolismo , Fatores de Transcrição Sp/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Transcrição Gênica/efeitos dos fármacos
13.
J Virol ; 81(13): 7274-9, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17428873

RESUMO

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.


Assuntos
Linfócitos B/metabolismo , Diferenciação Celular , Vírus da Leucemia Felina/metabolismo , Vírus da Leucemia Murina de Moloney/metabolismo , Células Progenitoras Mieloides/metabolismo , Infecções por Retroviridae/metabolismo , Infecções Tumorais por Vírus/mortalidade , Animais , Linfócitos B/patologia , Linfócitos B/virologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Medula Óssea/virologia , Gatos , Hematopoese Extramedular , Camundongos , Células Progenitoras Mieloides/patologia , Células Progenitoras Mieloides/virologia , Infecções por Retroviridae/patologia , Infecções Tumorais por Vírus/patologia
14.
Blood ; 108(12): 3691-9, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16931631

RESUMO

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.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus/fisiologia , Regulação Viral da Expressão Gênica/fisiologia , Células Progenitoras Mieloides/virologia , Ativação Viral/fisiologia , Latência Viral/fisiologia , Células Cultivadas , Técnicas de Cocultura , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/prevenção & controle , Fibroblastos/metabolismo , Fibroblastos/virologia , Perfilação da Expressão Gênica/métodos , Regulação Viral da Expressão Gênica/efeitos da radiação , Genoma Viral/fisiologia , Genoma Viral/efeitos da radiação , Humanos , Células Progenitoras Mieloides/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , RNA Viral/metabolismo , Transplante de Células-Tronco/efeitos adversos , Transplante de Células-Tronco/métodos , Raios Ultravioleta , Terapia Ultravioleta/métodos , Ativação Viral/efeitos da radiação , Inativação de Vírus/efeitos da radiação , Latência Viral/efeitos da radiação
15.
Methods Mol Biol ; 334: 199-209, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16861765

RESUMO

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.


Assuntos
Citomegalovirus/genética , Citomegalovirus/isolamento & purificação , DNA Viral/análise , DNA Viral/genética , Células Progenitoras Mieloides/virologia , Reação em Cadeia da Polimerase/métodos , Latência Viral/genética , Citomegalovirus/crescimento & desenvolvimento , Humanos , Hibridização In Situ , RNA Viral/análise , RNA Viral/genética
17.
Scand J Immunol ; 61(6): 545-50, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15963049

RESUMO

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.


Assuntos
Transplante de Medula Óssea , Vetores Genéticos , Células-Tronco Hematopoéticas/metabolismo , Linfócitos/metabolismo , Retroviridae/genética , Transdução Genética , Animais , Feminino , Citometria de Fluxo , Expressão Gênica , Células-Tronco Hematopoéticas/virologia , Proteínas Luminescentes/análise , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Linfócitos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/virologia , Baço/imunologia
18.
Res Vet Sci ; 78(2): 151-4, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15563922

RESUMO

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.


Assuntos
Doenças do Gato/virologia , Vírus da Leucemia Felina/genética , Vírus da Leucemia Felina/patogenicidade , Leucemia Mieloide Aguda/veterinária , Células Progenitoras Mieloides/virologia , Infecções por Retroviridae/veterinária , Sequências Repetidas Terminais , Infecções Tumorais por Vírus/veterinária , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/virologia , Gatos , Clonagem Molecular , DNA Viral/química , DNA Viral/genética , Leucemia Mieloide Aguda/patologia , Leucemia Mieloide Aguda/virologia , Síndromes Mielodisplásicas/veterinária , Síndromes Mielodisplásicas/virologia , Células Progenitoras Mieloides/citologia , Reação em Cadeia da Polimerase/veterinária , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia
19.
J Virol ; 78(8): 4054-62, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15047822

RESUMO

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.


Assuntos
Citomegalovirus/patogenicidade , Células Progenitoras Mieloides/fisiologia , Células Progenitoras Mieloides/virologia , Sequência de Bases , Células Cultivadas , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , DNA Complementar/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
J Immunol ; 171(10): 5571-8, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14607965

RESUMO

At least two distinct human dendritic cell (DC) subsets are produced in the bone marrow and circulate in the peripheral blood-precursor myeloid DCs (pre-mDCs) and plasmacytoid DCs (PDCs). Both lineages of DCs are instrumental in antiviral innate immunity and shaping Th1 adaptive immune responses. PDCs are the most potent IFN-alpha-producing cells to viral pathogens. Dengue, an acute flavivirus disease, provides a model to study DC responses to a self-limited human viral infection. We analyzed circulating DC subsets in a prospective study of children with dengue across a broad range of illness severities: healthy controls; mild, nondengue, presumed viral infections; moderately ill dengue fever; and, the most severe form of illness, dengue hemorrhagic fever. We also examined PDC responses in monkeys with asymptomatic dengue viremia and to dengue virus exposure in vitro. The absolute number and frequency of circulating pre-mDCs early in acute viral illness decreased as illness severity increased. Depressed pre-mDC blood levels appeared to be part of the typical innate immune response to acute viral infection. The frequency of circulating PDCs trended upward and the absolute number of circulating PDCs remained stable early in moderately ill children with dengue fever, mild other, nondengue, febrile illness, and monkeys with asymptomatic dengue viremia. However, there was an early decrease in circulating PDC levels in children who subsequently developed dengue hemorrhagic fever. A blunted blood PDC response to dengue virus infection was associated with higher viremia levels, and was part of an altered innate immune response and pathogenetic cascade leading to severe disease.


Assuntos
Células Dendríticas/imunologia , Dengue/imunologia , Plasmócitos/imunologia , Índice de Gravidade de Doença , Doença Aguda , Adolescente , Animais , Criança , Pré-Escolar , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Células Dendríticas/virologia , Dengue/tratamento farmacológico , Dengue/patologia , Modelos Animais de Doenças , Feminino , Humanos , Lactente , Interferon Tipo I/uso terapêutico , Interferon-alfa/metabolismo , Contagem de Leucócitos , Macaca mulatta , Masculino , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/patologia , Células Progenitoras Mieloides/virologia , Plasmócitos/metabolismo , Plasmócitos/patologia , Plasmócitos/virologia , Estudos Prospectivos , Proteínas Recombinantes , Dengue Grave/imunologia , Dengue Grave/patologia , Viremia/tratamento farmacológico , Viremia/imunologia , Viremia/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...