Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 309
Filtrar
1.
Retrovirology ; 21(1): 8, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38693565

RESUMO

The study of HIV infection and pathogenicity in physical reservoirs requires a biologically relevant model. The human immune system (HIS) mouse is an established model of HIV infection, but defects in immune tissue reconstitution remain a challenge for examining pathology in tissues. We utilized exogenous injection of the human recombinant FMS-like tyrosine kinase 3 ligand (rFLT-3 L) into the hematopoietic stem cell (HSC) cord blood HIS mouse model to significantly expand the total area of lymph node (LN) and the number of circulating human T cells. The results enabled visualization and quantification of HIV infectivity, CD4 T cell depletion and other measures of pathogenesis in the secondary lymphoid tissues of the spleen and LN. Treatment with the Caspase-1/4 inhibitor VX-765 limited CD4+ T cell loss in the spleen and reduced viral load in both the spleen and axillary LN. In situ hybridization further demonstrated a decrease in viral RNA in both the spleen and LN. Transcriptomic analysis revealed that in vivo inhibition of caspase-1/4 led to an upregulation in host HIV restriction factors including SAMHD1 and APOBEC3A. These findings highlight the use of rFLT-3 L to augment human immune system characteristics in HIS mice to support investigations of HIV pathogenesis and test host directed therapies, though further refinements are needed to further augment LN architecture and cellular populations. The results further provide in vivo evidence of the potential to target inflammasome pathways as an avenue of host-directed therapy to limit immune dysfunction and virus replication in tissue compartments of HIV+ persons.


Assuntos
Linfócitos T CD4-Positivos , Modelos Animais de Doenças , Infecções por HIV , HIV-1 , Animais , Camundongos , Infecções por HIV/imunologia , Infecções por HIV/virologia , Infecções por HIV/tratamento farmacológico , HIV-1/fisiologia , HIV-1/efeitos dos fármacos , Humanos , Linfócitos T CD4-Positivos/imunologia , Tecido Linfoide/virologia , Tecido Linfoide/imunologia , Carga Viral/efeitos dos fármacos , Baço/virologia , Baço/imunologia , Linfonodos/imunologia , Linfonodos/virologia , Caspases/metabolismo , Inibidores de Caspase/farmacologia , Antirretrovirais/uso terapêutico
2.
J Virol ; 98(2): e0165223, 2024 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-38299866

RESUMO

CCR5-tropic simian/human immunodeficiency viruses (SHIV) with clade C transmitted/founder envelopes represent a critical tool for the investigation of HIV experimental vaccines and microbicides in nonhuman primates, although many such isolates lead to spontaneous viral control post infection. Here, we generated a high-titer stock of pathogenic SHIV-C109p5 by serial passage in two rhesus macaques (RM) and tested its virulence in aged monkeys. The co-receptor usage was confirmed before infecting five geriatric rhesus macaques (four female and one male). Plasma viral loads were monitored by reverse transcriptase-quantitative PCR (RT-qPCR), cytokines by multiplex analysis, and biomarkers of gastrointestinal damage by enzyme-linked immunosorbent assay. Antibodies and cell-mediated responses were also measured. Viral dissemination into tissues was determined by RNAscope. Intravenous SHIV-C109p5 infection of aged RMs leads to high plasma viremia and rapid disease progression; rapid decrease in CD4+ T cells, CD4+CD8+ T cells, and plasmacytoid dendritic cells; and wasting necessitating euthanasia between 3 and 12 weeks post infection. Virus-specific cellular immune responses were detected only in the two monkeys that survived 4 weeks post infection. These were Gag-specific TNFα+CD8+, MIP1ß+CD4+, Env-specific IFN-γ+CD4+, and CD107a+ T cell responses. Four out of five monkeys had elevated intestinal fatty acid binding protein levels at the viral peak, while regenerating islet-derived protein 3α showed marked increases at later time points in the three animals surviving the longest, suggesting gut antimicrobial peptide production in response to microbial translocation post infection. Plasma levels of monocyte chemoattractant protein-1, interleukin-15, and interleukin-12/23 were also elevated. Viral replication in gut and secondary lymphoid tissues was extensive.IMPORTANCESimian/human immunodeficiency viruses (SHIV) are important reagents to study prevention of virus acquisition in nonhuman primate models of HIV infection, especially those representing transmitted/founder (T/F) viruses. However, many R5-tropic SHIV have limited fitness in vivo leading to many monkeys spontaneously controlling the virus post acute infection. Here, we report the generation of a pathogenic SHIV clade C T/F stock by in vivo passage leading to sustained viral load set points, a necessity to study pathogenicity. Unexpectedly, administration of this SHIV to elderly rhesus macaques led to extensive viral replication and fast disease progression, despite maintenance of a strict R5 tropism. Such age-dependent rapid disease progression had previously been reported for simian immunodeficiency virus but not for R5-tropic SHIV infections.


Assuntos
Infecções por HIV , HIV , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Replicação Viral , Animais , Feminino , Masculino , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Envelhecimento , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Quimiocina CCL2/imunologia , Quimiocina CCL2/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/patologia , Progressão da Doença , HIV/classificação , HIV/crescimento & desenvolvimento , HIV/patogenicidade , HIV/fisiologia , Infecções por HIV/imunologia , Infecções por HIV/patologia , Infecções por HIV/virologia , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucinas/imunologia , Interleucinas/metabolismo , Intestinos/virologia , Tecido Linfoide/virologia , Macaca mulatta/imunologia , Macaca mulatta/metabolismo , Inoculações Seriadas , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/classificação , Vírus da Imunodeficiência Símia/crescimento & desenvolvimento , Vírus da Imunodeficiência Símia/patogenicidade , Vírus da Imunodeficiência Símia/fisiologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Carga Viral , Tropismo Viral , Virulência , Receptores CCR5/metabolismo
3.
J Virol ; 97(6): e0176022, 2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37223960

RESUMO

CD4+ T follicular helper (TFH) cells are key targets for human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) replication and contribute to the virus reservoir under antiretroviral therapy (ART). Here, we describe a novel CD3+ CD20+ double-positive (DP) lymphocyte subset, resident in secondary lymphoid organs of humans and rhesus macaques (RMs), that appear predominantly after membrane exchange between TFH and B cells. DP lymphocytes are enriched in cells displaying a TFH phenotype (CD4+ PD1hi CXCR5hi), function (interleukin 21 positive [IL-21+]), and gene expression profile. Importantly, expression of CD40L upon brief in vitro mitogen stimulation identifies, by specific gene-expression signatures, DP cells of TFH-cell origin versus those of B-cell origin. Analysis of 56 RMs showed that DP cells (i) significantly increase following SIV infection, (ii) are reduced after 12 months of ART in comparison to pre-ART levels, and (iii) expand to a significantly higher frequency following ART interruption. Quantification of total SIV-gag DNA on sorted DP cells from chronically infected RMs showed that these cells are susceptible to SIV infection. These data reinforce earlier observations that CD20+ T cells are infected and expanded by HIV infection, while suggesting that these cells phenotypically overlap activated CD4+ TFH cells that acquire CD20 expression via trogocytosis and can be targeted as part of therapeutic strategies aimed at HIV remission. IMPORTANCE The HIV reservoir is largely composed of latently infected memory CD4+ T cells that persist during antiretroviral therapy and constitute a major barrier toward HIV eradication. In particular, CD4+ T follicular helper cells have been demonstrated as key targets for viral replication and persistence under ART. In lymph nodes from HIV-infected humans and SIV-infected rhesus macaques, we show that CD3+ CD20+ lymphocytes emerge after membrane exchange between T cells and B cells and are enriched in phenotypic, functional, and gene expression profiles found in T follicular helper cells. Furthermore, in SIV-infected rhesus macaques, these cells expand following experimental infection and after interruption of ART and harbor SIV DNA at levels similar to those found in CD4+ T cells; thus, CD3+ CD20+ lymphocytes are susceptible to SIV infection and can contribute to SIV persistence.


Assuntos
Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Células T Auxiliares Foliculares , Animais , Humanos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , Linfonodos/citologia , Macaca mulatta , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Células T Auxiliares Foliculares/imunologia , Células T Auxiliares Foliculares/virologia , Linfócitos B/imunologia , Linfócitos B/virologia , Ligante de CD40/genética , Expressão Gênica/imunologia , DNA Viral/metabolismo , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Tecido Linfoide/virologia
4.
Surg Pathol Clin ; 14(1): 75-96, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33526225

RESUMO

Lymphoepithelial carcinoma of salivary glands (LECSG) is an uncommon neoplasm. This article summarizes the findings of 438 cases in a review of the literature. Concurrent lymphoepithelial lesions may suggest a primary tumor. The tumor shows a nonkeratinizing carcinoma intimately associated with a rich lymphohistiocytic infiltrate, destroying adjacent salivary gland tissue. Irrespective of race or ethnicity, the tumors usually express Epstein-Barr virus, with Epstein-Barr virus encoded small RNA (EBER) and/or latent membrane protein-1 (LMP-1), although a subset does not. There is an overall good prognosis of about 80% at 5 years.


Assuntos
Carcinoma/patologia , Tecido Linfoide/patologia , Neoplasias das Glândulas Salivares/patologia , Carcinoma/epidemiologia , Carcinoma/cirurgia , Carcinoma/virologia , Diagnóstico Diferencial , Infecções por Vírus Epstein-Barr/epidemiologia , Infecções por Vírus Epstein-Barr/patologia , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/isolamento & purificação , Humanos , Imuno-Histoquímica , Hibridização In Situ , Incidência , Tecido Linfoide/virologia , Neoplasias Nasofaríngeas/diagnóstico , Reação em Cadeia da Polimerase , Neoplasias das Glândulas Salivares/epidemiologia , Neoplasias das Glândulas Salivares/cirurgia , Neoplasias das Glândulas Salivares/virologia
5.
J Virol ; 95(1)2020 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-33028711

RESUMO

Gammaherpesviruses (GHVs) are DNA tumor viruses that establish lifelong, chronic infections in lymphocytes of humans and other mammals. GHV infections are associated with numerous cancers, especially in immunocompromised hosts. While it is known that GHVs utilize host germinal center (GC) B cell responses during latency establishment, an understanding of how viral gene products function in specific B cell subsets to regulate this process is incomplete. Using murine gammaherpesvirus 68 (MHV68) as a small-animal model to define mechanisms of GHV pathogenesis in vivo, we generated a virus in which the M2 gene was flanked by loxP sites (M2.loxP), enabling the use of Cre-lox technology to define M2 function in specific cell types in infection and disease. The M2 gene encodes a protein that is highly expressed in GC B cells that promotes plasma cell differentiation and viral reactivation. M2 was efficiently deleted in Cre-expressing cells, and the presence of loxP sites flanking M2 did not alter viral replication or latency in mice that do not express Cre. In contrast, M2.loxP MHV68 exhibited a deficit in latency establishment and reactivation that resembled M2-null virus, following intranasal (IN) infection of mice that express Cre in all B cells (CD19-Cre). Nearly identical phenotypes were observed for M2.loxP MHV68 in mice that express Cre in germinal center (GC) B cells (AID-Cre). However, colonization of neither draining lymph nodes after IN infection nor the spleen after intraperitoneal (IP) infection required M2, although the reactivation defect was retained. Together, these data confirm that M2 function is B cell-specific and demonstrate that M2 primarily functions in AID-expressing cells to facilitate MHV68 dissemination to distal latency reservoirs within the host and reactivation from latency. Our study reveals that a viral latency gene functions within a distinct subset of cells to facilitate host colonization.IMPORTANCE Gammaherpesviruses establish lifelong chronic infections in cells of the immune system that can lead to lymphomas and other diseases. To facilitate colonization of a host, gammaherpesviruses encode gene products that manipulate processes involved in cellular proliferation and differentiation. Whether and how these viral gene products function in specific cells of the immune system is poorly defined. We report here the use of a viral genetic system that allows for deletion of specific viral genes in discrete populations of cells. We employ this system in an in vivo model to demonstrate cell-type-specific requirements for a particular viral gene. Our findings reveal that a viral gene product can function in distinct cellular subsets to direct gammaherpesvirus pathogenesis.


Assuntos
Linfócitos B/imunologia , Citidina Desaminase/imunologia , Infecções por Herpesviridae/virologia , Rhadinovirus/fisiologia , Proteínas Virais/imunologia , Ativação Viral , Animais , Antígenos CD19/metabolismo , Linfócitos B/virologia , Diferenciação Celular , Proliferação de Células , Centro Germinativo/imunologia , Centro Germinativo/virologia , Infecções por Herpesviridae/imunologia , Tecido Linfoide/imunologia , Tecido Linfoide/virologia , Camundongos , Rhadinovirus/genética , Rhadinovirus/metabolismo , Proteínas Virais/genética , Latência Viral
6.
Commun Biol ; 3(1): 376, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32665623

RESUMO

Viral replication is defined by the cellular microenvironment and one key factor is local oxygen tension, where hypoxia inducible factors (HIFs) regulate the cellular response to oxygen. Human immunodeficiency virus (HIV) infected cells within secondary lymphoid tissues exist in a low-oxygen or hypoxic environment in vivo. However, the majority of studies on HIV replication and latency are performed under laboratory conditions where HIFs are inactive. We show a role for HIF-2α in restricting HIV transcription via direct binding to the viral promoter. Hypoxia reduced tumor necrosis factor or histone deacetylase inhibitor, Romidepsin, mediated reactivation of HIV and inhibiting HIF signaling-pathways reversed this phenotype. Our data support a model where the low-oxygen environment of the lymph node may suppress HIV replication and promote latency. We identify a mechanism that may contribute to the limited efficacy of latency reversing agents in reactivating HIV and suggest new strategies to control latent HIV-1.


Assuntos
HIV-1/fisiologia , Latência Viral/fisiologia , Replicação Viral/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Microambiente Celular , Citometria de Fluxo , Humanos , Hipóxia/metabolismo , Hipóxia/virologia , Tecido Linfoide/metabolismo , Tecido Linfoide/virologia , Oxigênio , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase em Tempo Real , Transcrição Viral/fisiologia , Ativação Viral
7.
Proc Natl Acad Sci U S A ; 117(8): 4292-4299, 2020 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-32034098

RESUMO

The migratory patterns of virus-specific CD8 T cells during chronic viral infection are not well understood. To address this issue, we have done parabiosis experiments during chronic lymphocytic choriomeningitis virus (LCMV) infection of mice. We found that despite the high frequency of virus-specific CD8 T cells in both lymphoid and nonlymphoid tissues there was minimal migration of virus-specific CD8 T cells between the chronically infected conjoined parabiont mice. This was in contrast to parabionts between mice that had undergone an acute LCMV infection where virus-specific CD8 T cells established equilibrium demonstrating circulation of memory T cells generated after viral clearance. We have identified a population of PD-1+ TCF1+CXCR5+Tim-3- stemlike virus-specific CD8 T cells that reside in lymphoid tissues and act as resource cells for maintaining the T cell response during chronic infection. These are the cells that proliferate and give rise to the more terminally differentiated PD-1+ CXCR5-Tim-3+ CD8 T cells. Both the stemlike CD8 T cells and their terminally differentiated progeny showed minimal migration during chronic infection and the few LCMV-specific CD8 T cells that were present in circulation were the recently emerging progeny from the stemlike CD8 T cells. The PD-1+ TCF1+CXCR5+ stemlike CD8 T cells were truly resident in lymphoid tissues and did not circulate in the blood. We propose that this residency in specialized niches within lymphoid tissues is a key aspect of their biology and is essential for maintaining their quiescence and stemlike program under conditions of a chronic viral infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/fisiologia , Tecido Linfoide/imunologia , Receptor de Morte Celular Programada 1/imunologia , Animais , Feminino , Receptor Celular 2 do Vírus da Hepatite A/genética , Receptor Celular 2 do Vírus da Hepatite A/imunologia , Humanos , Memória Imunológica , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/genética , Tecido Linfoide/virologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/genética , Receptores CXCR5/genética , Receptores CXCR5/imunologia
8.
J Virol ; 93(24)2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31578287

RESUMO

CD137, a member of the tumor necrosis factor receptor superfamily of cell surface proteins, acts as a costimulatory receptor on T cells, natural killer cells, B cell subsets, and some dendritic cells. Agonistic anti-CD137 monoclonal antibody (MAb) therapy has been combined with other chemotherapeutic agents in human cancer trials. Based on its ability to promote tumor clearance, we hypothesized that anti-CD137 MAb might activate immune responses and resolve chronic viral infections. We evaluated anti-CD137 MAb therapy in a mouse infection model of chikungunya virus (CHIKV), an alphavirus that causes chronic polyarthritis in humans and is associated with reservoirs of CHIKV RNA that are not cleared efficiently by adaptive immune responses. Analysis of viral tropism revealed that CHIKV RNA was present preferentially in splenic B cells and follicular dendritic cells during the persistent phase of infection, and animals lacking B cells did not develop persistent CHIKV infection in lymphoid tissue. Anti-CD137 MAb treatment resulted in T cell-dependent clearance of CHIKV RNA in lymphoid tissue, although this effect was not observed in musculoskeletal tissue. The clearance of CHIKV RNA from lymphoid tissue by anti-CD137 MAb was associated with reductions in the numbers of germinal center B cells and follicular dendritic cells. Similar results were observed with anti-CD137 MAb treatment of mice infected with Mayaro virus, a related arthritogenic alphavirus. Thus, anti-CD137 MAb treatment promotes resolution of chronic alphavirus infection in lymphoid tissues by reducing the numbers of target cells for infection and persistence.IMPORTANCE Although CHIKV causes persistent infection in lymphoid and musculoskeletal tissues in multiple animals, the basis for this is poorly understood, which has hampered pharmacological efforts to promote viral clearance. Here, we evaluated the therapeutic effects on persistent CHIKV infection of an agonistic anti-CD137 MAb that can activate T cell and natural killer cell responses to clear tumors. We show that treatment with anti-CD137 MAb promotes the clearance of persistent alphavirus RNA from lymphoid but not musculoskeletal tissues. This occurs because anti-CD137 MAb-triggered T cells reduce the numbers of target germinal center B cells and follicular dendritic cells, which are the primary reservoirs for CHIKV in the spleen and lymph nodes. Our studies help to elucidate the basis for CHIKV persistence and begin to provide strategies that can clear long-term cellular reservoirs of infection.


Assuntos
Anticorpos Monoclonais/farmacologia , Febre de Chikungunya/imunologia , Vírus Chikungunya/efeitos dos fármacos , Tecido Linfoide/virologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Imunidade Adaptativa , Animais , Anticorpos Monoclonais/imunologia , Linfócitos B/imunologia , Febre de Chikungunya/virologia , Modelos Animais de Doenças , Humanos , Células Matadoras Naturais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Viral , Baço/virologia , Linfócitos T/imunologia , Tropismo Viral
9.
Front Immunol ; 10: 2109, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31552045

RESUMO

As the AIDS epidemic unfolded, the appearance of opportunistic infections in at-risk persons provided clues to the underlying problem: a dramatic defect in cell-mediated immunity associated with infection and depletion of CD4+ T lymphocytes. Moreover, the emergence of HIV-associated malignancies in these same individuals was a clear indication of the significant role effective cellular immunity plays in combating cancers. As research in the HIV field progressed, advances included the first demonstration of the role of PD-1 in human T cell exhaustion, and the development of gene-modified T cell therapies, including chimeric antigen receptor (CAR) T cells. In the intervening years, the oncology field has capitalized on these advances, effectively mobilizing the cellular immune response to achieve immune-mediated remission or cure of previously intractable cancers. Although similar therapeutic advances have not yet been achieved in the HIV field, spontaneous CD8+ T cell mediated remission or functional cure of HIV infection does occur in very small subset of individuals in the absence of anti-retroviral therapy (ART). This has many similarities to the CD8+ T cell mediated functional control or elimination of cancers, and indicates that immunotherapy for HIV is a rational goal. In HIV infection, one major barrier to successful immunotherapy is the small, persistent population of infected CD4+ T cells, the viral reservoir, which evades pharmacological and immune-mediated clearance, and is largely maintained in secondary lymphoid tissues at sites where CD8+ T cells have limited access and/or function. The reservoir-enriched lymphoid microenvironment bears a striking resemblance to the tumor microenvironment of many solid tumors-namely high levels of anti-inflammatory cytokines, expression of co-inhibitory receptors, and physical exclusion of immune effector cells. Here, we review the parallels between CD8+ T cell-mediated immune control of HIV and cancer, and how advances in cancer immunotherapy may provide insights to direct the development of effective HIV cure strategies. Specifically, understanding the impact of the tissue microenvironment on T cell function and development of CAR T cells and therapeutic vaccines deserve robust attention on the path toward a CD8+ T cell mediated cure of HIV infection.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por HIV/imunologia , HIV-1/fisiologia , Imunoterapia Adotiva/métodos , Tecido Linfoide/virologia , Neoplasias/imunologia , Animais , Linfócitos T CD4-Positivos/transplante , Infecções por HIV/terapia , Humanos , Tolerância Imunológica , Imunidade Celular , Neoplasias/terapia , Receptor de Morte Celular Programada 1/metabolismo , Remissão Espontânea , Microambiente Tumoral , Latência Viral
10.
Vaccine ; 37(43): 6397-6404, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31515142

RESUMO

BACKGROUND: Marek's disease (MD) is a lymphoproliferative disease of chickens caused by Marek's disease virus (MDV), an oncogenic α-herpesvirus. Since 1970, MD has been controlled by widespread vaccination; however, more effective MD vaccines are needed to counter more virulent MDV strains. The bivalent vaccine combination of SB-1 and herpesvirus of turkey (HVT) strain FC126 has been widely used. Nonetheless, the mechanism(s) underlying this synergistic effect has not been investigated. METHODS: Three experiments were conducted where SB-1 or HVT were administered as monovalent or bivalent vaccines to newly hatched chickens, then challenged five days later with MDV. In Experiment 1, levels of MDV replication in PBMCs were measured over time, and tumor incidence and vaccinal protection determined. In Experiment 2, MDV and vaccine strains replication levels in lymphoid organs were measured at 1, 5, 10, and 14 days post-challenge (DPC). In Experiment 3, to verify that the bursa was necessary for HVT protection, a subset of chicks were bursectomized and these birds plus controls were similarly vaccinated and challenged, and the levels of vaccinal protection determined. RESULTS: The efficacy of bivalent SB-1 + HVT surpasses that of either SB-1 or HVT monovalent vaccines in controlling the level of pathogenic MDV in PBMCs until the end of the study, and this correlated with the ability to inhibit tumor formation. SB-1 replication in the spleen increased from 1 to 14 DPC, while HVT replicated only in the bursa at 1 DPC. The bursa was necessary for immune protection induced by HVT vaccine. CONCLUSION: Synergy of SB-1 and HVT vaccines is due to additive influences of the individual vaccines acting at different times and target organs. And the bursa is vital for HVT to replicate and induce immune protection.


Assuntos
Linfoma/veterinária , Vacinas contra Doença de Marek/imunologia , Doenças das Aves Domésticas/prevenção & controle , Vacinação/veterinária , Vacinas Virais/imunologia , Animais , Galinhas/imunologia , Sinergismo Farmacológico , Herpesvirus Meleagrídeo 1/imunologia , Herpesvirus Galináceo 2/imunologia , Herpesvirus Galináceo 2/fisiologia , Leucócitos Mononucleares/virologia , Tecido Linfoide/virologia , Linfoma/prevenção & controle , Linfoma/virologia , Vacinas contra Doença de Marek/administração & dosagem , Cavidade Peritoneal/virologia , Doenças das Aves Domésticas/virologia , Vacinas Virais/administração & dosagem , Replicação Viral
11.
Cell Host Microbe ; 26(3): 347-358.e7, 2019 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-31471273

RESUMO

Viral rebound upon stopping combined antiretroviral therapy poses a major barrier toward an HIV cure. Cellular and anatomical sources responsible for reinitiating viral replication remain a subject of ardent debate, despite extensive research efforts. To unravel the source of rebounding viruses, we conducted a large-scale HIV-STAR (HIV-1 sequencing before analytical treatment interruption to identify the anatomically relevant HIV reservoir) clinical trial. We collected samples from 11 participants and compared the genetic composition of (pro)viruses collected under treatment from different cellular and anatomical compartments with that of plasma viruses sampled during analytical treatment interruption. We found a remarkably heterogeneous source of viral rebound. In addition, irrespective of the compartment or cell subset, genetically identical viral expansions played a significant role in viral rebound. Our study suggests that although there does not seem to be a primary source for rebound HIV, cellular proliferation is an important driver of HIV persistence and should therefore be considered in future curative strategies.


Assuntos
Infecções por HIV/virologia , HIV-1/genética , Dispositivos de Acesso Vascular/virologia , Antirretrovirais/uso terapêutico , Medula Óssea/virologia , Proliferação de Células , Líquido Cefalorraquidiano/virologia , Feminino , Genes Virais , HIV-1/isolamento & purificação , Humanos , Cinética , Linfonodos/virologia , Tecido Linfoide/virologia , Masculino , Plasma , Carga Viral , Replicação Viral
12.
Virologie (Montrouge) ; 23(4): 211-228, 2019 08 01.
Artigo em Francês | MEDLINE | ID: mdl-31414659

RESUMO

Current antiretroviral therapy allows the control of HIV replication but a relapse occurs most of the time in case of treatment interruption. The viral genome integration explains this persistence of HIV in all body tissues, at very variable levels depending on their density of CD4+ T-cells, HIV main target. Secondary lymphoid tissues are the most infected organs. Several techniques can be used to characterize the reservoir, detecting different forms of the virus. They are complementary to decipher the establishment of HIV reservoir during the primary infection and its dynamics afterwards. In peripheral blood, the earlier the initiation of treatment, the more important is the decrease in total HIV DNA. Early treatment prevents the progressive increase in stable integrated forms of HIV DNA and preserves immune cells from infection. A better understanding of HIV infection in controllers will also aid in the development of new therapeutic strategies targeting the reservoir.


Assuntos
Reservatórios de Doenças/virologia , Infecções por HIV/virologia , HIV-1/fisiologia , Tecido Linfoide/virologia , Tropismo Viral/fisiologia , Fármacos Anti-HIV/farmacologia , Fármacos Anti-HIV/uso terapêutico , Biomarcadores , Células Cultivadas , DNA Viral/análise , Células Dendríticas/virologia , Progressão da Doença , Sistemas de Liberação de Medicamentos , Infecções por HIV/tratamento farmacológico , Humanos , Linfócitos/virologia , Macrófagos/virologia , Monócitos/virologia , Provírus/genética , Provírus/fisiologia , Transcrição Gênica , Integração Viral , Latência Viral , Replicação Viral
13.
Vet Microbiol ; 235: 136-142, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31282371

RESUMO

Infectious Bursal Disease Virus (IBDV) of the ITA genotype (G6) was shown to have peculiar molecular characteristics and, despite a subclinical course, aggressiveness towards lymphoid tissues after experimental infection of specific-pathogen-free (SPF) chickens. The aim of the present study was to evaluate and compare with a Classical IBDV strain, ITA IBDV distribution and persistence in various tissues (bursa of Fabricious, spleen, thymus, bone marrow, caecal tonsils, Harderian gland, kidney, liver and proventriculus), its cloacal shedding and the involvement of gut TLR-3 in duodenum tissues. The 35-day-old SPF chickens were experimentally infected and sampled up to 28 days post infection (dpi) for IBDV detection and TLR-3 quantification by qRT-PCR. The ITA IBDV strain was detected in lymphoid and most non-lymphoid tissues up to the end of the trial, with higher loads compared to the Classical IBDV. Most of those differences were found during the first 2 weeks post-infection. Notably, bone marrow and caecal tonsils presented higher viral loads until 28 dpi, allowing to speculate that these organs may serve as non-bursal lymphoid tissues supporting virus replication. Differences in relative TLR-3 gene expression between ITA IBDV-infected birds and Classical-IBDV infected ones were observed at 4, 14 and 21 dpi, being initially higher in Classical group and later in ITA group. Our results provide new insights into IBDV pathogenesis showing that IBDV of ITA genotype leads to a high and persistent viral load in lymphoid tissues and to a delayed antiviral response.


Assuntos
Infecções por Birnaviridae/veterinária , Vírus da Doença Infecciosa da Bursa/genética , Tecido Linfoide/virologia , Doenças das Aves Domésticas/imunologia , Carga Viral , Animais , Infecções por Birnaviridae/imunologia , Medula Óssea/patologia , Medula Óssea/virologia , Galinhas , Ensaio de Imunoadsorção Enzimática , Genótipo , Vírus da Doença Infecciosa da Bursa/patogenicidade , Tonsila Palatina/virologia , Doenças das Aves Domésticas/virologia , Organismos Livres de Patógenos Específicos , Receptor 3 Toll-Like/genética , Replicação Viral
14.
Vet Microbiol ; 235: 164-169, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31282374

RESUMO

Marek's disease virus (MDV) is a highly contagious alphaherpesvirus that causes rapid onset of T cell lymphomas in chickens. MDV continues to break through vaccinal immunity due to the emergence of highly virulent field strains. Earlier studies revealed that deletion of the meq gene from MDV results in attenuated vaccines that protect against disease when chickens are infected with highly virulent strains. However, meq-deleted viruses still retain the ability to induce lymphoid organ atrophy, which raises safety concerns. In an earlier study, we found that deletion of lorf9 counteracts this lymphoid organ atrophy. Here, we describe the generation of a double deletion mutant virus lacking virus-encoded meq and lorf9. In vitro studies revealed that during replication, the mutant virus had kinetic characteristics similar to the parental virus; however, in vivo the replication capability was significantly reduced. Results of animal studies revealed no obvious MDV-specific symptoms and lesions. Importantly, the double deletion mutant virus lost the capacity to induce lymphoid organ atrophy, which has been the main obstacle during development of a good vaccine candidate.


Assuntos
Deleção de Genes , Herpesvirus Galináceo 1/genética , Herpesvirus Galináceo 1/patogenicidade , Tecido Linfoide/patologia , Doença de Marek/patologia , Proteínas Oncogênicas Virais/genética , Animais , Atrofia , Galinhas , Tecido Linfoide/virologia , Mutação , Doenças das Aves Domésticas/virologia , Replicação Viral
15.
Retrovirology ; 15(1): 68, 2018 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-30292240

RESUMO

Traditionally, NK cells belong to the innate immune system and eliminate virus-infected cells through their germline-encoded receptors. However, NK cells were recently reported to possess memory-like functions that were predominantly provided by hepatic NK cells. Memory properties were mainly documented in contact hypersensitivity models or during cytomegalovirus infections. However, the precise role and the physiologic importance of memory-like NK cells during retroviral infections are still under investigation. Here, we show that Friend retrovirus (FV) infection of mice induced a population of phenotypically memory-like NK cells at 28 days post infection. Upon secondary antigen encounter, these NK cells showed an increased production of the pro-inflammatory cytokines IFNγ and TNFα as well as the death ligand FasL in comparison to naïve NK cells. Furthermore, we found an augmented elimination of antigen-matched but not antigen-mismatched target cells by these memory-like NK cells. In adoptive cell transfer experiments, equal antiviral activities of splenic and hepatic memory-like NK cells during the late phase of acute FV infection were found. Our results strongly imply the existence and antiviral activity of spleen and liver memory-like NK cells in FV infection, which efficiently respond upon secondary exposure to retroviral antigens.


Assuntos
Vírus da Leucemia Murina de Friend/fisiologia , Memória Imunológica , Células Matadoras Naturais/imunologia , Infecções por Retroviridae/imunologia , Transferência Adotiva , Animais , Antígenos Virais/imunologia , Feminino , Vírus da Leucemia Murina de Friend/imunologia , Células Matadoras Naturais/citologia , Tecido Linfoide/imunologia , Tecido Linfoide/virologia , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo
16.
Mucosal Immunol ; 11(5): 1342-1351, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29875402

RESUMO

Human gut-associated lymphoid tissues (GALT) play a key role in the acute phase of HIV infection. The propensity of HIV to replicate in these tissues, however, is not fully understood. Access and migration of naive and memory CD4+ T cells to these sites is mediated by interactions between integrin α4ß7, expressed on CD4+ T cells, and MAdCAM, expressed on high endothelial venules. We report here that MAdCAM delivers a potent costimulatory signal to naive and memory CD4+ T cells following ligation with α4ß7. Such costimulation promotes high levels of HIV replication. An anti-α4ß7 mAb that prevents mucosal transmission of SIV blocks MAdCAM signaling through α4ß7 and MAdCAM-dependent viral replication. MAdCAM costimulation of memory CD4+ T cells is sufficient to drive cellular proliferation and the upregulation of CCR5, while naive CD4+ T cells require both MAdCAM and retinoic acid to achieve the same response. The pairing of MAdCAM and retinoic acid is unique to the GALT, leading us to propose that HIV replication in these sites is facilitated by MAdCAM-α4ß7 interactions. Moreover, complete inhibition of MAdCAM signaling by an anti-α4ß7 mAb, an analog of the clinically approved therapeutic vedolizumab, highlights the potential of such agents to control acute HIV infection.


Assuntos
Infecções por HIV/metabolismo , HIV/fisiologia , Integrinas/metabolismo , Replicação Viral/fisiologia , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Feminino , HIV/efeitos dos fármacos , Infecções por HIV/tratamento farmacológico , Humanos , Memória Imunológica/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/virologia , Tecido Linfoide/metabolismo , Tecido Linfoide/virologia , Macaca mulatta , Domínios e Motivos de Interação entre Proteínas , Receptores CCR5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tretinoína/metabolismo , Regulação para Cima/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
17.
Hum Pathol ; 79: 18-41, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29885408

RESUMO

Epstein-Barr virus (EBV) has been linked to many human neoplasms including hematopoietic, epithelial, and mesenchymal tumors. Since our original review of EBV-associated lymphoproliferative disorders in 2007, many advances and developments have been reported. In this review, we will examine the recent advances in EBV-associated lymphoid/histiocytic proliferations, dividing them into reactive, B cell, T/NK cell, immunodeficiency-related, and histiocytic/dendritic cell proliferations.


Assuntos
Proliferação de Células , Infecções por Vírus Epstein-Barr/patologia , Herpesvirus Humano 4/patogenicidade , Tecido Linfoide/patologia , Transtornos Linfoproliferativos/patologia , Infecções por Vírus Epstein-Barr/virologia , Humanos , Tecido Linfoide/virologia , Transtornos Linfoproliferativos/virologia , Prognóstico
18.
Antivir Ther ; 23(6): 495-504, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29714167

RESUMO

BACKGROUND: Raltegravir (RTG) and dolutegravir (DTG) have different pharmacokinetic patterns in the gastrointestinal tract. To determine if this results in pharmacodynamic differences, we compared HIV RNA, HIV DNA and immunological markers in gut-associated lymphoid tissue (GALT) of HIV-infected participants receiving RTG or DTG with tenofovir+emtricitabine (TDF/FTC). METHODS: GALT specimens from the terminal ileum, splenic flexure and rectum were obtained by colonoscopy at a single time point in 20 adults treated with RTG (n=10) or DTG (n=10) with HIV RNA <50 copies/ml. Flow cytometry, drug concentrations, and HIV RNA and DNA were analysed in tissue. CD4/8+ T-cells were tested for γδ TCR, and markers of T-cell activation and exhaustion. Data are reported as median (Q1-Q3). RESULTS: A total of 15 men and 5 women were enrolled. There was no difference in time since HIV diagnosis for those on RTG (9.5 [4-22] years) and DTG (17 [1-24] years; P=0.6), although time on RTG (5.4 [2.3-6.7] years) was greater than DTG (1.0 [0.1-1.5] years; P<0.001). Concentrations of RTG and DTG in rectal tissue were similar to previous reports: median tissue:plasma ratio was 11.25 for RTG and 0.44 for DTG. RNA:DNA ratios were 1.14 (0.18-5.10) for the RTG group and 0.90 (0.30-18.87) for the DTG group (P=0.95). No differences (P≥0.1) between CD4+ and CD8+ T-cell markers were found. CONCLUSIONS: RTG produced higher tissue exposures than DTG, but no significant differences in GALT HIV RNA, DNA or most immunological markers were observed. ClinicalTrials.gov NCT02218320.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Compostos Heterocíclicos com 3 Anéis/uso terapêutico , Tecido Linfoide/efeitos dos fármacos , Raltegravir Potássico/uso terapêutico , Adulto , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/virologia , Colo Transverso/efeitos dos fármacos , Colo Transverso/patologia , Colo Transverso/virologia , DNA Viral/antagonistas & inibidores , DNA Viral/genética , DNA Viral/metabolismo , Emtricitabina/uso terapêutico , Feminino , Expressão Gênica , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/genética , HIV-1/imunologia , Humanos , Íleo/efeitos dos fármacos , Íleo/patologia , Íleo/virologia , Imunidade Inata/efeitos dos fármacos , Tecido Linfoide/patologia , Tecido Linfoide/virologia , Masculino , Pessoa de Meia-Idade , Oxazinas , Piperazinas , Piridonas , RNA Viral/antagonistas & inibidores , RNA Viral/genética , RNA Viral/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/genética , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Reto/efeitos dos fármacos , Reto/patologia , Reto/virologia , Tenofovir/uso terapêutico , Resultado do Tratamento
19.
PLoS Pathog ; 14(2): e1006856, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29470552

RESUMO

HIV-1-infected cells persist indefinitely despite the use of combination antiretroviral therapy (ART), and novel therapeutic strategies to target and purge residual infected cells in individuals on ART are urgently needed. Here, we demonstrate that CD4+ T cell-associated HIV-1 RNA is often highly enriched in cells expressing CD30, and that cells expressing this marker considerably contribute to the total pool of transcriptionally active CD4+ lymphocytes in individuals on suppressive ART. Using in situ RNA hybridization studies, we show co-localization of CD30 with HIV-1 transcriptional activity in gut-associated lymphoid tissues. We also demonstrate that ex vivo treatment with brentuximab vedotin, an antibody-drug conjugate (ADC) that targets CD30, significantly reduces the total amount of HIV-1 DNA in peripheral blood mononuclear cells obtained from infected, ART-suppressed individuals. Finally, we observed that an HIV-1-infected individual, who received repeated brentuximab vedotin infusions for lymphoma, had no detectable virus in peripheral blood mononuclear cells. Overall, CD30 may be a marker of residual, transcriptionally active HIV-1 infected cells in the setting of suppressive ART. Given that CD30 is only expressed on a small number of total mononuclear cells, it is a potential therapeutic target of persistent HIV-1 infection.


Assuntos
Linfócitos T CD4-Positivos/virologia , Infecções por HIV/virologia , HIV-1/fisiologia , Antígeno Ki-1/metabolismo , Tecido Linfoide/virologia , Reto/virologia , Ativação Transcricional , Fármacos Anti-HIV/farmacologia , Terapia Antirretroviral de Alta Atividade , Biomarcadores/sangue , Biomarcadores/metabolismo , Brentuximab Vedotin , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Células Cultivadas , Estudos de Coortes , DNA Viral/sangue , DNA Viral/metabolismo , Infecções por HIV/tratamento farmacológico , Infecções por HIV/metabolismo , Infecções por HIV/patologia , HIV-1/efeitos dos fármacos , HIV-1/isolamento & purificação , HIV-1/patogenicidade , Humanos , Imunoconjugados/farmacologia , Hibridização In Situ , Antígeno Ki-1/antagonistas & inibidores , Antígeno Ki-1/sangue , Antígeno Ki-1/química , Tecido Linfoide/efeitos dos fármacos , Tecido Linfoide/metabolismo , Tecido Linfoide/patologia , RNA Viral/sangue , RNA Viral/metabolismo , Reto/efeitos dos fármacos , Reto/metabolismo , Reto/patologia , Solubilidade , Ativação Transcricional/efeitos dos fármacos , Carga Viral/efeitos dos fármacos
20.
J Neurovirol ; 24(1): 1-15, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29063512

RESUMO

HIV-1 Nef is a flexible, multifunctional protein with several cellular targets that is required for pathogenicity of the virus. This protein maintains a high degree of genetic variation among intra- and inter-host isolates. HIV Nef is relevant to HIV-associated neurological diseases (HAND) in patients treated with combined antiretroviral therapy because of the protein's role in promoting survival and migration of infected brain macrophages. In this study, we analyzed 2020 HIV Nef sequences derived from 22 different tissues and 31 subjects using a novel computational approach. This approach combines statistical regression and evolved neural networks (ENNs) to classify brain sequences based on the physical and chemical characteristics of functional Nef domains. Based on training, testing, and validation data, the method successfully classified brain Nef sequences at 84.5% and provided informative features for further examination. These included physicochemical features associated with the Src-homology-3 binding domain, the Nef loop (including the AP-2 Binding region), and a cytokine-binding domain. Non-brain sequences from patients with HIV-associated neurological disease were frequently classified as brain, suggesting that the approach could indicate neurological risk using blood-derived virus or for the development of biomarkers for use in assay systems aimed at drug efficacy studies for the treatment of HIV-associated neurological diseases.


Assuntos
Complexo AIDS Demência/virologia , Encéfalo/virologia , HIV-1/genética , Interações Hospedeiro-Patógeno/genética , Produtos do Gene nef do Vírus da Imunodeficiência Humana/química , Complexo AIDS Demência/tratamento farmacológico , Complexo AIDS Demência/genética , Complexo AIDS Demência/fisiopatologia , Sequência de Aminoácidos , Fármacos Anti-HIV/uso terapêutico , Terapia Antirretroviral de Alta Atividade , Autopsia , Sítios de Ligação , Encéfalo/metabolismo , Encéfalo/patologia , Expressão Gênica , HIV-1/metabolismo , HIV-1/patogenicidade , Humanos , Tecido Linfoide/metabolismo , Tecido Linfoide/patologia , Tecido Linfoide/virologia , Macrófagos/metabolismo , Macrófagos/patologia , Macrófagos/virologia , Modelos Moleculares , Redes Neurais de Computação , Especificidade de Órgãos , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA