Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Eur J Immunol ; 52(3): 447-461, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34935145

RESUMO

Effective function of CD8+ T cells and enhanced innate activation of DCs in response to HIV-1 is linked to protective antiviral immunity in controllers. Manipulation of DC targeting the master regulator TANK-binding Kinase 1 (TBK1) might be useful to acquire controller-like properties. Here, we evaluated the impact of the combination of 2´3´-c´diAM(PS)2 and Poly I:C as potential adjuvants capable of potentiating DC´s abilities to induce polyfunctional HIV-1 specific CD8+ T-cell responses in vitro and in vivo using a humanized BLT mouse model. Adjuvant combination enhanced TBK-1 phosphorylation and IL-12 and IFN-ß expression on DC and increased their ability to activate polyfunctional HIV-1-specific CD8+ T cells in vitro. Moreover, higher proportions of hBLT mice vaccinated with ADJ-DC exhibited less severe CD4+ T-cell depletion following HIV-1 infection compared to control groups. This was associated with infiltration of CD8+ T cells in the white pulp from the spleen, reduced spread of infected p24+ cells to LN, and with preserved abilities of CD8+ T cells from the spleen and blood of vaccinated animals to induce specific polyfunctional responses upon antigen stimulation. Therefore, priming of DC with PolyI:C and STING agonists might be useful for future HIV-1 vaccine studies.


Assuntos
Vacinas contra a AIDS , HIV-1 , Vacinas contra a AIDS/metabolismo , Adjuvantes Imunológicos/farmacologia , Animais , Linfócitos T CD4-Positivos , Linfócitos T CD8-Positivos , Células Dendríticas , Proteína do Núcleo p24 do HIV/metabolismo , Tecido Linfoide , Camundongos , Poli I-C/farmacologia
3.
Cell Rep ; 36(9): 109622, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34469717

RESUMO

HIV-1 entry into host cells leads to one of the following three alternative fates: (1) HIV-1 elimination by restriction factors, (2) establishment of HIV-1 latency, or (3) active viral replication in target cells. Here, we report the development of an improved system for monitoring HIV-1 fate at single-cell and population levels and show the diverse applications of this system to study specific aspects of HIV-1 fate in different cell types and under different environments. An analysis of the transcriptome of infected, primary CD4+ T cells that support alternative fates of HIV-1 identifies differential gene expression signatures in these cells. Small molecules are able to selectively target cells that support viral replication with no significant effect on viral latency. In addition, HIV-1 fate varies in different tissues following infection of humanized mice in vivo. Altogether, these studies indicate that intra- and extra-cellular environments contribute to the fate of HIV-1 infection.


Assuntos
Linfócitos T CD4-Positivos/virologia , Microambiente Celular , Infecções por HIV/virologia , HIV-1/patogenicidade , Animais , Fármacos Anti-HIV/farmacologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Células HEK293 , Infecções por HIV/tratamento farmacológico , Infecções por HIV/genética , Infecções por HIV/imunologia , HIV-1/efeitos dos fármacos , HIV-1/crescimento & desenvolvimento , HIV-1/imunologia , Interações Hospedeiro-Patógeno , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Células THP-1 , Transcriptoma , Internalização do Vírus , Latência Viral , Replicação Viral
4.
J Biol Chem ; 294(29): 11199-11212, 2019 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-31167786

RESUMO

Tick evasins (EVAs) bind either CC- or CXC-chemokines by a poorly understood promiscuous or "one-to-many" mechanism to neutralize inflammation. Because EVAs potently inhibit inflammation in many preclinical models, highlighting their potential as biological therapeutics for inflammatory diseases, we sought to further unravel the CXC-chemokine-EVA interactions. Using yeast surface display, we identified and characterized 27 novel CXC-chemokine-binding evasins homologous to EVA3 and defined two functional classes. The first, which included EVA3, exclusively bound ELR+ CXC-chemokines, whereas the second class bound both ELR+ and ELR- CXC-chemokines, in several cases including CXC-motif chemokine ligand 10 (CXCL10) but, surprisingly, not CXCL8. The X-ray crystal structure of EVA3 at a resolution of 1.79 Å revealed a single antiparallel ß-sheet with six conserved cysteine residues forming a disulfide-bonded knottin scaffold that creates a contiguous solvent-accessible surface. Swapping analyses identified distinct knottin scaffold segments necessary for different CXC-chemokine-binding activities, implying that differential ligand positioning, at least in part, plays a role in promiscuous binding. Swapping segments also transferred chemokine-binding activity, resulting in a hybrid EVA with dual CXCL10- and CXCL8-binding activities. The solvent-accessible surfaces of the knottin scaffold segments have distinctive shape and charge, which we suggest drives chemokine-binding specificity. These studies provide structural and mechanistic insight into how CXC-chemokine-binding tick EVAs achieve class specificity but also engage in promiscuous binding.


Assuntos
Quimiocinas CXC/metabolismo , Miniproteínas Nó de Cistina/metabolismo , Receptores de Quimiocinas/metabolismo , Carrapatos/metabolismo , Animais , Cristalografia por Raios X , Ligação Proteica , Conformação Proteica , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/isolamento & purificação , Especificidade da Espécie , Carrapatos/classificação , Leveduras/genética
5.
Cell Rep ; 26(6): 1409-1418.e5, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30726727

RESUMO

Lung interstitial CD4+ T cells are critical for protection against pulmonary infections, but the fate of this population during HIV-1 infection is not well described. We studied CD4+ T cells in the setting of HIV-1 infection in human lung tissue, humanized mice, and a Mycobacterium tuberculosis (Mtb)/simian immunodeficiency virus (SIV) nonhuman primate co-infection model. Infection with a CCR5-tropic strain of HIV-1 or SIV results in severe and rapid loss of lung interstitial CD4+ T cells but not blood or lung alveolar CD4+ T cells. This is accompanied by high HIV-1 production in these cells in vitro and in vivo. Importantly, during early SIV infection, loss of lung interstitial CD4+ T cells is associated with increased dissemination of pulmonary Mtb infection. We show that lung interstitial CD4+ T cells serve as an efficient target for HIV-1 and SIV infection that leads to their early depletion and an increased risk of disseminated tuberculosis.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Coinfecção/imunologia , Infecções por HIV/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Tuberculose Pulmonar/imunologia , Animais , Linfócitos T CD4-Positivos/patologia , Coinfecção/patologia , Feminino , Células HEK293 , Infecções por HIV/patologia , HIV-1/patogenicidade , Humanos , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Pulmão/virologia , Macaca mulatta , Camundongos , Mycobacterium tuberculosis/patogenicidade , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Vírus da Imunodeficiência Símia/patogenicidade , Tuberculose Pulmonar/patologia
6.
Nat Biotechnol ; 37(3): 293-302, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30742125

RESUMO

Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative treatment for multiple disorders, but deficiency and dysregulation of T cells limit its utility. Here we report a biomaterial-based scaffold that mimics features of T cell lymphopoiesis in the bone marrow. The bone marrow cryogel (BMC) releases bone morphogenetic protein-2 to recruit stromal cells and presents the Notch ligand Delta-like ligand-4 to facilitate T cell lineage specification of mouse and human hematopoietic progenitor cells. BMCs subcutaneously injected in mice at the time of HSCT enhanced T cell progenitor seeding of the thymus, T cell neogenesis and diversification of the T cell receptor repertoire. Peripheral T cell reconstitution increased ~6-fold in mouse HSCT and ~2-fold in human xenogeneic HSCT. Furthermore, BMCs promoted donor CD4+ regulatory T cell generation and improved survival after allogeneic HSCT. In comparison to adoptive transfer of T cell progenitors, BMCs increased donor chimerism, T cell generation and antigen-specific T cell responses to vaccination. BMCs may provide an off-the-shelf approach for enhancing T cell regeneration and mitigating graft-versus-host disease in HSCT.


Assuntos
Transplante de Medula Óssea , Doença Enxerto-Hospedeiro/imunologia , Transplante de Células-Tronco Hematopoéticas , Linfócitos T Reguladores/imunologia , Alicerces Teciduais , Transferência Adotiva/métodos , Animais , Medula Óssea , Quimerismo , Doença Enxerto-Hospedeiro/patologia , Doença Enxerto-Hospedeiro/terapia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Camundongos , Linfócitos T Reguladores/citologia , Transplante Heterólogo/métodos , Transplante Homólogo
7.
Cell Host Microbe ; 25(1): 73-86.e5, 2019 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-30629922

RESUMO

HIV-1 primarily infects T lymphocytes and uses these motile cells as migratory vehicles for effective dissemination in the host. Paradoxically, the virus at the same time disrupts multiple cellular processes underlying lymphocyte motility, seemingly counterproductive to rapid systemic infection. Here we show by intravital microscopy in humanized mice that perturbation of the actin cytoskeleton via the lentiviral protein Nef, and not changes to chemokine receptor expression or function, is the dominant cause of dysregulated infected T cell motility in lymphoid tissue by preventing stable cellular polarization required for fast migration. Accordingly, disrupting the Nef hydrophobic patch that facilitates actin cytoskeletal perturbation initially accelerates systemic viral dissemination after female genital transmission. However, the same feature of Nef was subsequently critical for viral persistence in immune-competent hosts. Therefore, a highly conserved activity of lentiviral Nef proteins has dual effects and imposes both fitness costs and benefits on the virus at different stages of infection.


Assuntos
Citoesqueleto de Actina/metabolismo , Movimento Celular , Infecções por HIV/transmissão , HIV-1/fisiologia , HIV-1/patogenicidade , Mucosa/metabolismo , Actinas/metabolismo , Animais , Quimiocinas/metabolismo , Modelos Animais de Doenças , Feminino , Células HEK293 , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/imunologia , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Humanos , Linfócitos/virologia , Camundongos , Mucosa/virologia , Linfócitos T/imunologia , Linfócitos T/virologia , Proteínas Virais Reguladoras e Acessórias/metabolismo , Viremia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/imunologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Quinases Ativadas por p21/metabolismo
8.
Curr Opin Virol ; 25: 97-104, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28810166

RESUMO

Antiretroviral therapy can efficiently control HIV viral replication, resulting in low viral loads and sustained CD4+ T cell counts in HIV-infected persons. However, fast viral rebound occurs in most infected persons when therapy is interrupted. The principal component of persistent infection is a latent but replication-competent HIV reservoir. The long half-life of this reservoir is a major barrier to cure, and its elimination is the target of important research efforts. Animal models that can recapitulate this aspect of human infection are needed to examine the HIV reservoir in tissues in vivo, and to test eradication strategies. In this review, we will summarize recent studies using humanized mouse models to examine different aspects of the viral reservoir.


Assuntos
Modelos Animais de Doenças , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/fisiologia , Camundongos Transgênicos , Latência Viral , Animais , HIV-1/imunologia , Humanos , Camundongos , Carga Viral , Replicação Viral
9.
JCI Insight ; 2(7): e88533, 2017 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-28405607

RESUMO

HIV vaginal transmission accounts for the majority of newly acquired heterosexual infections. However, the mechanism by which HIV spreads from the initial site of viral entry at the mucosal surface of the female genital tract to establish a systemic infection of lymphoid and peripheral tissues is not known. Once the virus exits the mucosa it rapidly spreads to all tissues, leading to CD4+ T cell depletion and the establishment of a viral reservoir that cannot be eliminated with current treatments. Understanding the molecular and cellular requirements for viral dissemination from the genital tract is therefore of great importance, as it could reveal new strategies to lengthen the window of opportunity to target the virus at its entry site in the mucosa where it is the most vulnerable and thus prevent systemic infection. Using HIV vaginal infection of humanized mice as a model of heterosexual transmission, we demonstrate that blocking the ability of leukocytes to respond to chemoattractants prevented HIV from leaving the female genital tract. Furthermore, blocking lymphocyte egress from lymph nodes prevented viremia and infection of the gut. Leukocyte trafficking therefore plays a major role in viral dissemination, and targeting the chemoattractant molecules involved can prevent the establishment of a systemic infection.


Assuntos
Linfócitos T CD4-Positivos/virologia , Infecções por HIV/transmissão , HIV-1/patogenicidade , Receptores CCR7/imunologia , Receptores de Lisoesfingolipídeo/imunologia , Vagina/virologia , Animais , Linfócitos T CD4-Positivos/imunologia , Movimento Celular , Quimiocina CCL19/imunologia , Quimiocina CCL21/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Leucócitos/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores de Esfingosina-1-Fosfato , Vagina/imunologia
10.
J Infect Dis ; 214(4): 612-6, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27357340

RESUMO

Humanized mice reconstituted with a human immune system can be mucosally infected with human immunodeficiency virus (HIV), opening up the possibility of studying HIV transmission in a small-animal model. Here we report that passive immunization with the broadly neutralizing antibody b12 protected humanized mice against repetitive intravaginal infection in a dose-dependent manner. In addition, treatment with the antibody PGT126, which is more potent in vitro, was more efficacious in vivo and provided sterilizing protection. Our results demonstrate that humanized mice can be used as a small-animal model to study the efficacy and mechanism of broadly neutralizing antibody protection against HIV acquisition.


Assuntos
Anticorpos Neutralizantes/administração & dosagem , Modelos Animais de Doenças , Anticorpos Anti-HIV/administração & dosagem , Infecções por HIV/prevenção & controle , Imunização Passiva/métodos , Animais , Relação Dose-Resposta Imunológica , Feminino , Camundongos , Camundongos SCID , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA