Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Intervalo de año de publicación
1.
J Immunol ; 207(5): 1310-1321, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34380652

RESUMEN

The respiratory tract is constantly exposed to various airborne pathogens. Most vaccines against respiratory infections are designed for the parenteral routes of administration; consequently, they provide relatively minimal protection in the respiratory tract. A vaccination strategy that aims to induce the protective mucosal immune responses in the airway is urgently needed. The FcRn mediates IgG Ab transport across the epithelial cells lining the respiratory tract. By mimicking this natural IgG transfer, we tested whether FcRn delivers vaccine Ags to induce a protective immunity to respiratory infections. In this study, we designed a monomeric IgG Fc fused to influenza virus hemagglutinin (HA) Ag with a trimerization domain. The soluble trimeric HA-Fc were characterized by their binding with conformation-dependent HA Abs or FcRn. In wild-type, but not FcRn knockout, mice, intranasal immunization with HA-Fc plus CpG adjuvant conferred significant protection against lethal intranasal challenge with influenza A/PR/8/34 virus. Further, mice immunized with a mutant HA-Fc lacking FcRn binding sites or HA alone succumbed to lethal infection. Protection was attributed to high levels of neutralizing Abs, robust and long-lasting B and T cell responses, the presence of lung-resident memory T cells and bone marrow plasma cells, and a remarkable reduction of virus-induced lung inflammation. Our results demonstrate for the first time, to our knowledge, that FcRn can effectively deliver a trimeric viral vaccine Ag in the respiratory tract and elicit potent protection against respiratory infection. This study further supports a view that FcRn-mediated mucosal immunization is a platform for vaccine delivery against common respiratory pathogens.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Infecciones por Orthomyxoviridae/inmunología , Orthomyxoviridae/fisiología , Receptores Fc/metabolismo , Mucosa Respiratoria/metabolismo , Administración Intranasal , Animales , Anticuerpos Antivirales/metabolismo , Modelos Animales de Enfermedad , Resistencia a la Enfermedad , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/metabolismo , Vacunas contra la Influenza/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Fc/genética , Mucosa Respiratoria/inmunología , Vacunación
2.
J Immunol ; 197(5): 1884-92, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27474072

RESUMEN

A successful pregnancy depends on the maintenance of tolerance at the fetal-maternal interface; strong inflammation in the placental bed is generally associated with adverse fetal outcomes. Among the mechanisms that foster tolerance and limit inflammation, the fetal immune system favors Th2 or regulatory responses over Th1 responses. The unintended consequence of this functional program is high susceptibility to infections. Human Vδ2 T cells mount innate-like responses to a broad range of microorganisms and are poised for Th1 responses before birth. In infants they likely play a key role in protection against pathogens by exerting early Th1 effector functions, improving function of other innate cells, and promoting Th1 polarization of adaptive responses. However, their propensity to release Th1 mediators may require careful regulation during fetal life to avoid exaggerated proinflammatory responses. We investigated molecules with the potential to act as a rheostat for fetal Vδ2 cells. Programmed death 1 (PD1) is a negative regulator of T cell responses and a determinant of tolerance, particularly at the fetal-maternal interface. Neonatal Vδ2 cells upregulate PD1 shortly after activation and, unlike their adult counterparts, express this molecule for at least 28 d. Engagement of PD1 by one of its ligands, PDL1, effectively dampens TCR-mediated responses (TNF-α production and degranulation) by neonatal Vδ2 cells and may thus help maintain their activity within safe limits. PD1 expression by neonatal Vδ2 cells is inversely associated with promoter DNA methylation. Prolonged PD1 expression may be part of a functional program to control Vδ2 cell inflammatory responses during fetal life.


Asunto(s)
Epigénesis Genética , Regulación de la Expresión Génica , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Antígeno B7-H1/inmunología , Metilación de ADN , Femenino , Desarrollo Fetal , Humanos , Tolerancia Inmunológica , Recién Nacido , Inflamación/inmunología , Activación de Linfocitos , Embarazo , Regiones Promotoras Genéticas , Células TH1/inmunología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/inmunología
3.
Immunology ; 151(4): 385-394, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28342224

RESUMEN

The study aimed to identify an immunoregulatory factor that restores the phosphoantigen response of Vγ9Vδ2+ T cells from HIV-positive individuals on antiretroviral therapy. It was designed to characterize the effects of interleukin-18 (IL-18) on proliferation and effector function in Vγ9Vδ2 T cells from HIV-negative individuals and test whether exogenous IL-18 reconstitutes the Vγ9Vδ2 T-cell response to phosphoantigen from HIV-positive donors. Vγ9Vδ2 T cells from HIV-negative individuals responded strongly to phosphoantigen or aminobisphosphonate stimulation of peripheral blood mononuclear cells (PBMC), whereas cells with similar T-cell receptor profiles from HIV-positive individuals only responded to aminobisphosphonate. Interleukin-18 was higher after aminobisphosphonate stimulation due to activation of the inflammasome pathway. Both IL-18 and IL-18 receptor levels were measured and the activity of exogenous IL-18 on HIV-negative and HIV-positive PBMC was evaluated in terms of Vγ9Vδ2 T-cell proliferation, memory subsets, cytokine expression and CD107a expression. Interleukin-18 stimulation increased proliferation, enhanced the accumulation of effector memory cells, and increased expression of cytotoxic markers in HIV-negative controls. When Vγ9Vδ2 T cells from HIV-positive individuals were stimulated with isopentenyl pyrophosphate in the presence of IL-18, there was increased proliferation, accumulation of memory cells, and higher expression of CD56, NKG2D and CD107a (markers of cytotoxic effector phenotype). Interleukin-18 stimulation specifically expanded the Vγ9-JγP+ subset of Vγ9Vδ2 T cells, as was expected for normal responses to phosphoantigen. Interleukin-18 is a potent stimulator of Vγ9Vδ2 T-cell proliferation and effector function. Therapies directed at reconstituting Vγ9Vδ2 T-cell activity in HIV-positive individuals should include stimulators of IL-18 or direct cytokine supplementation.


Asunto(s)
Infecciones por VIH/inmunología , VIH-1/inmunología , Inmunoterapia/métodos , Interleucina-18/metabolismo , Células Asesinas Naturales/inmunología , Linfocitos T/inmunología , Antígenos/inmunología , Proliferación Celular , Células Cultivadas , Difosfonatos/metabolismo , Hemiterpenos/inmunología , Humanos , Imidazoles/metabolismo , Memoria Inmunológica , Inmunofenotipificación , Activación de Linfocitos , Compuestos Organofosforados/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/virología , Ácido Zoledrónico
4.
Eur J Immunol ; 45(1): 298-308, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25263533

RESUMEN

The majority of CXCR5(+) PD1(+) CD4(+) T follicular helper (Tfh) cells (>90%) are CD25(-) Bcl6(hi) , while a small subpopulation (<10%) are CD25(+) Bcl6(low) but do not express FoxP3 and are not T regulatory cells. We purified T:B-cell conjugates from tonsils and found they were enriched for the CD25(+) Bcl6(low) Tfh-cell subpopulation. In response to IL-2, these CD25(+) Tfh cells increased expression of costimulatory molecules ICOS or OX40, upregulated transcription factor cMaf, produced cytokines IL-21, IL-17, and IL-10, and raised the levels of antiapoptotic protein Bcl2. Conjugates formed with CD25(+) BCl6(low) Tfh cells included B cells expressing higher levels of activation-induced cytidine deaminase (AID), memory marker CD45RO, surface IgG or IgA, and MHC class II compared to B-cell conjugates including CD25(-) Bcl6(hi) Tfh cells. While IL-2 suppresses early Tfh-cell differentiation, Tfh-cell recognition of antigen-presenting B cells and signaling through the T-cell receptor likely triggers expression of the high-affinity IL-2 receptor and responses to IL-2 including downregulation of Bcl6. CD25 expression on Tfh cells and local production of IL-2 in tonsil or lymph node may support B helper T-cell function during later stages of B-cell maturation and the development of immune memory.


Asunto(s)
Linfocitos B/citología , Proteínas de Unión al ADN/genética , Centro Germinal/citología , Tonsila Palatina/citología , Receptores de Interleucina-2/genética , Linfocitos T Colaboradores-Inductores/citología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Diferenciación Celular , Citidina Desaminasa/genética , Citidina Desaminasa/inmunología , Citocinas/genética , Citocinas/inmunología , Proteínas de Unión al ADN/inmunología , Regulación de la Expresión Génica , Centro Germinal/efectos de los fármacos , Centro Germinal/inmunología , Humanos , Memoria Inmunológica , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Interleucina-2/farmacología , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/inmunología , Activación de Linfocitos/efectos de los fármacos , Tonsila Palatina/efectos de los fármacos , Tonsila Palatina/inmunología , Proteínas Proto-Oncogénicas c-bcl-6 , Proteínas Proto-Oncogénicas c-maf/genética , Proteínas Proto-Oncogénicas c-maf/inmunología , Receptores de Interleucina-2/inmunología , Receptores OX40/genética , Receptores OX40/inmunología , Transducción de Señal , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología
5.
Cell Immunol ; 296(1): 22-30, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25769734

RESUMEN

Lymphocytes expressing a T cell receptor (TCR) composed of Vgamma9 and Vdelta2 chains represent a minor fraction of human thymocytes. Extrathymic selection throughout post-natal life causes the proportion of cells with a Vgamma9-JP rearrangement to increase and elevates the capacity for responding to non-peptidic phosphoantigens. Extrathymic selection is so powerful that phosphoantigen-reactive cells comprise about 1 in 40 circulating memory T cells in healthy adults and the subset expands rapidly upon infection or in response to malignancy. Skewing of the gamma delta TCR repertoire is accompanied by selection for public gamma chain sequences such that many unrelated individuals overlap extensive in their circulating repertoire. This type of selection implies the presence of a monomorphic antigen-presenting molecule that is an object of current research but remains incompletely defined. While selection on a monomorphic presenting molecule may seem unusual, similar mechanisms shape the alpha beta T cell repertoire including the extreme examples of NKT or mucosal-associated invariant T cells (MAIT) and the less dramatic amplification of public Vbeta chain rearrangements driven by individual MHC molecules and associated with resistance to viral pathogens. Selecting and amplifying public T cell receptors whether alpha beta or gamma delta, are important steps in developing an anticipatory TCR repertoire. Cell clones expressing public TCR can accelerate the kinetics of response to pathogens and impact host survival.


Asunto(s)
Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Subgrupos de Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Evolución Molecular , Humanos , Memoria Inmunológica/inmunología , Células T Asesinas Naturales/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Homología de Secuencia , Timocitos/inmunología
6.
J Infect Dis ; 209(10): 1653-62, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24325967

RESUMEN

BACKGROUND: Plasmodium falciparum placental infection primes the fetal immune system and alters infant immunity. Mechanisms leading to these outcomes are not completely understood. We focused on Vγ2Vδ2 cells, which are part of the immune response against many pathogens, including P. falciparum. These unconventional lymphocytes respond directly to small, nonpeptidic antigens, independent of major histocompatibility complex presentation. We wondered whether placental malaria, which may increase fetal exposure to P. falciparum metabolites, triggers a response by neonatal Vγ2Vδ2 lymphocytes that can be a marker for the extent of fetal exposure to malarial antigens. METHODS: Cord blood mononuclear cells were collected from 15 neonates born to mothers with P. falciparum infection during pregnancy (8 with placental malaria) and 25 unexposed neonates. Vγ2Vδ2 cell phenotype, repertoire, and proliferative responses were compared between newborns exposed and those unexposed to P. falciparum. RESULTS: Placental malaria-exposed neonates had increased proportions of central memory Vγ2Vδ2 cells in cord blood, with an altered Vγ2 chain repertoire ex vivo and after stimulation. CONCLUSION: Our results suggest that placental malaria affects the phenotype and repertoire of neonatal Vγ2Vδ2 lymphocytes. Placental malaria may lower the capacity for subsequent Vγ2Vδ2 cell responses and impair the natural resistance to infectious diseases or the response to pediatric vaccination.


Asunto(s)
Sangre Fetal/citología , Inmunidad Materno-Adquirida , Malaria Falciparum/inmunología , Complicaciones Parasitarias del Embarazo/inmunología , Subgrupos de Linfocitos T/fisiología , Biomarcadores , Femenino , Regulación de la Expresión Génica/inmunología , Humanos , Cadenas gamma de Inmunoglobulina/genética , Cadenas gamma de Inmunoglobulina/metabolismo , Recién Nacido , Embarazo
7.
Immunology ; 2014 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-25227493

RESUMEN

Interactions between NK and dendritic cells (DC) affect maturation and function of both cell populations, including NK killing of DC (editing) that is important for controlling the quality of immune responses. We also know that antigen-stimulated Vγ2Vδ2 T cells costimulate NK cells via 4-1BB to enhance killing of tumor cell lines but we do not know what regulates 4-1BB expression or whether other NK effector functions including DC killing, might also be influenced by NK:γδ T cell cross talk. Here we show that antigen-stimulated γδ T cells costimulate NK through ICOS:ICOSL and this signal increases NK killing of autologous DC. Effects of NK:γδ T cell co-culture, which could be reproduced with soluble ICOS-Fc fusion protein, included increased CD69 and 4-1BB expression, IFN-γ, TNF-α, MIP-1ß, I-309, RANTES and sFasL production, as well as elevated mRNA levels for costimulatory receptors OX40 (TNFRSF4) and GITR (TNFRSF18). Thus, ICOS/ICOSL costimulation of NK by Vγ2Vδ2 T cells had broad effects on NK phenotype and effector functions. The NK γδ T cell cross talk links innate and antigen-specific lymphocyte responses in the control of cytotoxic effector function and dendritic cell killing. This article is protected by copyright. All rights reserved.

8.
Immunology ; 138(4): 346-60, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23181340

RESUMEN

Compared with adults, the circulating Vγ2Vδ2 T-cell population in cord blood is present at low levels and does not show the strong bias for Vγ2-Jγ1.2 rearrangements. These features may be a result of limited exposure to stimulatory phosphoantigens, lack of T-cell-derived interleukin-2 (IL-2) or both. In cord blood mononuclear cell cultures, a single round of stimulation, using aminobisphosphonates to elevate phosphoantigen levels, resulted in expansion of adult-like Vγ2 chains and accumulation of memory cells with cytotoxic potential. Selection was similar using IL-2 or myeloid-derived IL-15. The Vγ2Vδ2 T cells present in neonates are capable of generating potent immune responses even when relying on IL-15.


Asunto(s)
Alendronato/farmacología , Sangre Fetal/efectos de los fármacos , Interleucina-15/farmacología , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Linfocitos T Citotóxicos/efectos de los fármacos , Adulto , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Sangre Fetal/citología , Sangre Fetal/inmunología , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Humanos , Interleucina-2/farmacología , Activación de Linfocitos/efectos de los fármacos , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Proteínas Recombinantes/farmacología , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología
9.
Retrovirology ; 10: 60, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23742646

RESUMEN

BACKGROUND: The cell surface receptors CD4 and CCR5 bind CCR5-tropic HIV Envelope (Env) glycoprotein during virus attachment. These same receptors have signaling activities related to normal immune cell functions. We also know that Env binds to CCR5 present at high levels on CD4-negative γδ T cells where it signals through p38 MAP kinase to activate caspases and Fas-independent cell death. Here, we asked whether Env signaling through cellular receptors is responsible for death among uninfected CD4+/CCR5+ T cells and what are the effects of Env on CD4+/CCR5-negative cells that might impact HIV infection. The outcomes of Env binding are analyzed in terms of signal transduction and the effects on cell activation or cell death pathways. RESULTS: Env binding to CD4 signals through Erk and Akt kinases. Activation of Erk/Akt suppresses p38 due to CCR5 binding, and allows cell survival. When CD4 signaling was blocked by soluble CD4 or protein kinase inhibitors, p38 activation and Fas-independent cell death were increased among uninfected CD4+ CCR5+ T cells. We also noted specific effects of CD4 signaling on CCR5-negative CD4 T cells in tonsil lymphocyte cultures. Exposure to CCR5-tropic HIV Env (BaL strain) increased expression of CXCR5, PD-1, Fas and FasL. Among CD4+/CCR5- T cells expressing high levels of CXCR5 and PD-1, there were substantial amounts of Fas-dependent cell death. Increased CXCR5 and PD-1 expression was blocked by soluble CD4 or specific inhibitors of the Akt kinase, showing a direct relationship between CD4 signaling, T cell activation and Fas-dependent cell death. CONCLUSIONS: Specific inhibition of Akt activation increased Env-dependent cell death of CCR5+ CD4 T cells. The same inhibitor, antibodies blocking the CD4 binding site on gp120, or soluble CD4 also prevented the increase in expression of CXCR5 or PD-1, and reduced the levels of Fas-dependent cell death. The Akt kinase and related signaling events, are key to cell survival that is needed for productive infection, and may be targets for the development of antivirals. Specific inhibitors of Akt would decrease productive infection, by favoring cell death during virus attachment to CD4+ CCR5+ target cells, and reduce immune activation to prevent Fas-dependent death of uninfected CXCR5+ PD-1+ CD4 T cells including T follicular helper cells that share this phenotype.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Muerte Celular , VIH/patogenicidad , Proteína Oncogénica v-akt/metabolismo , Factores de Virulencia/metabolismo , Productos del Gen env del Virus de la Inmunodeficiencia Humana/metabolismo , Antígenos CD4/metabolismo , Humanos , Depleción Linfocítica , Proteína Oncogénica v-akt/antagonistas & inhibidores , Receptores CCR5/metabolismo , Transducción de Señal
10.
Blood ; 118(22): 5824-31, 2011 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21926353

RESUMEN

HIV infects and replicates in CD4+ T cells but effects on host immunity and disease also involve depletion, hyper-activation, and modification of CD4-negative cell populations. In particular, the depletion of CD4-negative γδ T cells is common to all HIV+ individuals. We found that soluble or cell-associated envelope glycoproteins from CCR5-tropic strains of HIV could bind, activates the p38-caspase pathway, and induce the death of γδ cells. Envelope binding requires integrin α4ß7 and chemokine receptor CCR5 which are at high levels and form a complex on the γδ T cell membrane. This receptor complex facilitated V3 loop binding to CCR5 in the absence of CD4-induced conformational changes. Cell death was increased by antigen stimulation after exposure to envelope glycoprotein. Direct signaling by envelope glycoprotein killed CD4-negative γδ T cells and reproduced a defect observed in all patients with HIV disease.


Asunto(s)
Síndrome de Inmunodeficiencia Adquirida/inmunología , Apoptosis/efectos de los fármacos , Infecciones por VIH/inmunología , Integrinas/fisiología , Receptores CCR5/fisiología , Linfocitos T/efectos de los fármacos , Proteínas del Envoltorio Viral/farmacología , Síndrome de Inmunodeficiencia Adquirida/metabolismo , Apoptosis/fisiología , Antígenos CD4/metabolismo , Células Cultivadas , Progresión de la Enfermedad , VIH/genética , VIH/fisiología , VIH/ultraestructura , Proteína gp120 de Envoltorio del VIH/farmacología , Proteína gp120 de Envoltorio del VIH/fisiología , Infecciones por VIH/patología , Células HeLa , Humanos , Integrinas/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores CCR5/metabolismo , Linfocitos T/metabolismo , Linfocitos T/fisiología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/fisiología
11.
Virol J ; 10: 52, 2013 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-23402317

RESUMEN

BACKGROUND: Lassa hemorrhagic fever (LHF) is a rodent-borne viral disease that can be fatal for human beings. In this study, an attenuated Lassa vaccine candidate, ML29, was tested in SIV-infected rhesus macaques for its ability to elicit immune responses without instigating signs pathognomonic for arenavirus disease. ML29 is a reassortant between Lassa and Mopeia viruses that causes a transient infection in non-human primates and confers sterilizing protection from lethal Lassa viral challenge. However, since the LHF endemic area of West Africa also has high HIV seroprevalence, it is important to determine whether vaccination could be safe in the context of HIV infection. RESULTS: SIV-infected and uninfected rhesus macaques were vaccinated with the ML29 virus and monitored for specific humoral and cellular immune responses, as well as for classical and non-classical signs of arenavirus disease. Classical disease signs included viremia, rash, respiratory distress, malaise, high liver enzyme levels, and virus invasion of the central nervous system. Non-classical signs, derived from profiling the blood transcriptome of virulent and non-virulent arenavirus infections, included increased expression of interferon-stimulated genes (ISG) and decreased expression of COX2, IL-1ß, coagulation intermediates and nuclear receptors needed for stress signaling. All vaccinated monkeys showed ML29-specific antibody responses and ML29-specific cell-mediated immunity. CONCLUSION: SIV-infected and uninfected rhesus macaques responded similarly to ML29 vaccination, and none developed chronic arenavirus infection. Importantly, none of the macaques developed signs, classical or non-classical, of arenavirus disease.


Asunto(s)
Coinfección/inmunología , Infecciones por VIH/inmunología , Fiebre de Lassa/prevención & control , Virus Lassa/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/inmunología , Coinfección/prevención & control , Coinfección/virología , Infecciones por VIH/complicaciones , Infecciones por VIH/virología , Humanos , Fiebre de Lassa/complicaciones , Fiebre de Lassa/inmunología , Fiebre de Lassa/virología , Macaca mulatta , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas Virales/administración & dosificación
12.
J Infect Dis ; 205(9): 1448-55, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22454465

RESUMEN

BACKGROUND: CD4- Vγ2Vδ2 T cells are depleted during human immunodeficiency virus (HIV) infection but can recover to near normal levels in patients who spontaneously control viremia in the absence of therapy. By contrasting Vγ2Vδ2 T-cell numbers, phenotype, and T-cell receptor (TCR) repertoire, we investigate the dynamic tension between active immunity and progressive T-cell destruction during persistent viremia. METHODS: Peripheral blood Vγ2Vδ2 T-cell levels and phenotypes were characterized by flow cytometry. Lymphoproliferation assays measured functional responses. Spectratyping characterized damage to the TCR repertoire. RESULTS: Levels, responses to antigen and the proportion of T effector memory Vγ2Vδ2 T cells in patients with persistent viremia, were intermediate between patients with natural virus suppression (NVS) and patients receiving antiretroviral therapy. Damage to the TCR γ-2 chain repertoire and depletion of CD56+ Vγ2Vδ2 T cells were more pronounced in viremic patients, compared with antiretroviral therapy recipients and patients with natural virus suppression. CONCLUSIONS: Characteristics of Vγ2Vδ2 T cells in viremic patients reflect both active responses (increasing cell numbers, better antigen responses, and higher proportion of effector memory cells) and ongoing damage (repertoire changes and loss of CD56+ cells). Unlike patients who control viremia to undetectable levels, Vγ2Vδ2 T cells are diminished during persistent viremia and may eventually be lost because of progressive destruction of the TCR repertoire.


Asunto(s)
Infecciones por VIH/inmunología , VIH/fisiología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Replicación Viral , Adulto , Femenino , Infecciones por VIH/virología , Humanos , Masculino , Persona de Mediana Edad , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Estudios Retrospectivos , Linfocitos T/virología , Viremia/inmunología
13.
J Virol ; 85(20): 10542-53, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21849464

RESUMEN

Strategies to prevent the sexual transmission of HIV include vaccines that elicit durable, protective mucosal immune responses. A key to effective mucosal immunity is the capacity for antigens administered locally to cross epithelial barriers. Given the role of neonatal Fc receptor (FcRn) in transferring IgG across polarized epithelial cells which line mucosal surfaces, FcRn might be useful for delivering HIV vaccine antigens across mucosal epithelial barriers to the underlying antigen-presenting cells. Chimeric proteins composed of HIV Gag (p24) fused to the Fc region of IgG (Gag-Fc) bind efficiently to airway mucosa and are transported across this epithelial surface. Mice immunized intranasally with Gag-Fc plus CpG adjuvant developed local and systemic immunity, including durable B and T cell memory. Gag-specific immunity was sufficiently potent to protect against an intravaginal challenge with recombinant vaccinia virus expressing the HIV Gag protein. Intranasal administration of a Gag-Fc/CpG vaccine protected at a distal mucosal site. Our data suggest that targeting of FcRn with chimeric immunogens may be an important strategy for mucosal immunization and should be considered a new approach for preventive HIV vaccines.


Asunto(s)
Vacunas contra el SIDA/inmunología , Proteína p24 del Núcleo del VIH/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Receptores Fc/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Adyuvantes Inmunológicos/administración & dosificación , Administración Intranasal , Animales , Linfocitos B/inmunología , Femenino , Proteína p24 del Núcleo del VIH/genética , Infecciones por VIH/prevención & control , Fragmentos Fc de Inmunoglobulinas/genética , Memoria Inmunológica , Ratones , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
14.
Blood ; 116(10): 1726-33, 2010 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-20519625

RESUMEN

Natural killer (NK) cells are innate effector lymphocytes that control the growth of major histocompatibility complex class I negative tumors. We show here that γδ T lymphocytes, expanded in vitro in the presence isopentenylpyrophosphate (IPP), induce NK cell-mediated killing of tumors that are usually resistant to NK cytolysis. The induction of cytotoxicity toward these resistant tumors requires priming of NK cells by immobilized human immunoglobulin G1 and costimulation through CD137L expressed on activated γδ T lymphocytes. This costimulation increases NKG2D expression on the NK-cell surface, which is directly responsible for tumor cell lysis. Moreover, culturing peripheral blood mononuclear cells with zoledronic acid, a γδ T lymphocyte activating agent, enhances NK-cell direct cytotoxicity and antibody-dependent cellular cytotoxicity against hematopoietic and nonhematopoietic tumors. Our data reveal a novel function of human γδ T lymphocytes in the regulation of NK cell-mediated cytotoxicity and provide rationale for the use of strategies to manipulate the CD137 pathway to augment innate antitumor immunity.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Células Asesinas Naturales/inmunología , Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Ligando 4-1BB/inmunología , Ligando 4-1BB/metabolismo , Antígeno B7-1/inmunología , Antígeno B7-1/metabolismo , Antígeno B7-2/inmunología , Antígeno B7-2/metabolismo , Comunicación Celular/inmunología , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Cultura , Pruebas Inmunológicas de Citotoxicidad , Citometría de Flujo , Humanos , Inmunoglobulina G/inmunología , Células K562 , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Ligando OX40/inmunología , Ligando OX40/metabolismo , Unión Proteica , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
15.
Cytotherapy ; 14(2): 173-81, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22029653

RESUMEN

BACKGROUND AIMS: Immunotherapy using γδ T cells capable of mediating antibody-dependent cellular cytotoxicity (ADCC) is a promising anti-human immunodeficiency virus (HIV) strategy. Approved aminobispohsphonate drugs, for example zoledronate (Zometa), stimulate γδ T cells in cancer patients, where they may promote direct tumor killing. Knowing that γδ T cells are modulated during HIV disease, documenting their responses and potential for controlling HIV is important. We investigated whether zoledronate/interleukin (IL)-2 could expand cytotoxic Vδ2 cells from HIV+ donors and whether these cells functioned in ADCC. METHODS: Peripheral blood mononuclear cells from uninfected controls and HIV+ individuals receiving anti-retroviral therapy were treated with isopentenyl pyrophosphate (IPP) or zoledronate plus IL-2 to expand the Vδ2+ subset. Immunophenotyping and functional analyzes (cytotoxicity or cytokine expression) allowed us to compare cell properties from individual donors and to compare the responses to each stimulating agent. RESULTS: Zoledronate stimulated a greater expansion of Vδ2 cells in HIV+ individuals compared with phosphoantigen IPP, and these cells expressed CD16. CD56 expression (a marker for cytotoxic cells) was lower on zoledronate-expanded cells, consistent with significantly lower cytotoxicity against the Daudi tumor cell line. Cells expanded with either zoledronate or IPP were active in ADCC, were similar in terms of interferon (IFN)-γ and tumor necrosis factor (TNF)-α expression, and degranulated in response to Fc receptor cross-linking. CONCLUSIONS: Zoledronate causes ex vivo expansion of Vδ2 cells from HIV+ individuals. Despite lower expression of CD56 and decreased direct cytotoxicity, these effectors were potent in ADCC. Zoledronate/IL-2- expanded cells have potential for immunotherapy to activate Vδ2 cells in HIV patients and enhance ADCC.


Asunto(s)
Antirretrovirales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Difosfonatos/farmacología , Infecciones por VIH/inmunología , Imidazoles/farmacología , Interleucina-2/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Antígeno CD56/inmunología , Antígeno CD56/metabolismo , Línea Celular Tumoral , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , VIH/inmunología , Infecciones por VIH/terapia , Hemiterpenos/farmacología , Humanos , Inmunofenotipificación , Interleucina-2/metabolismo , Activación de Linfocitos/efectos de los fármacos , Compuestos Organofosforados/farmacología , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Ácido Zoledrónico
16.
Front Immunol ; 13: 1012051, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36275712

RESUMEN

Human Vγ9Vδ2 T cells are attractive candidates for cancer immunotherapy due to their potent capacity for tumor recognition and cytolysis of many tumor cell types. However, efforts to deploy clinical strategies for Vγ9Vδ2 T cell cancer therapy are hampered by insufficient potency. We are pursuing an alternate strategy of modifying tumors to increase the capacity for Vγ9Vδ2 T cell activation, as a means for strengthening the anti-tumor response by resident or ex vivo manufactured Vγ9Vδ2 T cells. Vγ9Vδ2 T cells are activated in vitro by non-peptidic antigens including isopentenyl pyrophosphate (IPP), a substrate of farnesyl diphosphate synthase (FDPS) in the pathway for biosynthesis of isoprenoids. In an effort to improve in vivo potency of Vγ9Vδ2 T cells, we reduced FDPS expression in tumor cells using a lentivirus vector encoding a short-hairpin RNA that targets FDPS mRNA (LV-shFDPS). Prostate (PC3) or hepatocellular carcinoma (Huh-7) cells transduced with LV-shFDPS induced Vγ9Vδ2 T cell stimulation in vitro, resulting in increased cytokine expression and tumor cell cytotoxicity. Immune deficient mice implanted with LV-shFDPS transduced tumor cells showed dramatic responses to intraperitoneal injection of Vγ9Vδ2 T cells with strong suppression of tumor growth. In vivo potency was increased by transducing tumor cells with a vector expressing both shFDPS and human IL-2. Tumor suppression by Vγ9Vδ2 T cells was dose-dependent with greater effects observed in mice injected with 100% LV-shFDPS transduced cells compared to mice injected with a mixture of 50% LV-shFDPS transduced cells and 50% control (no vector) tumor cells. Delivery of LV-shFDPS by intratumoral injection was insufficient to knockdown FDPS in the majority of tumor cells, resulting in insignificant tumor suppression by Vγ9Vδ2 T cells. Thus, Vγ9Vδ2 T cells efficiently targeted and suppressed tumors expressing shFDPS in mouse xenotransplant models. This proof-of-concept study demonstrates the potential for suppression of genetically modified tumors by human Vγ9Vδ2 T cells and indicates that co-expression of cytokines may boost the anti-tumor effect.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Masculino , Humanos , Ratones , Animales , Linfocitos T , Geraniltranstransferasa/genética , Geraniltranstransferasa/farmacología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Interleucina-2/farmacología , Xenoinjertos , ARN Mensajero , ARN
17.
Front Med (Lausanne) ; 9: 1044713, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36452901

RESUMEN

The cell and gene therapy product AGT103-T was designed to restore the Gag-specific CD4+ T cell response in persons with chronic HIV disease who are receiving antiretroviral therapy. This autologous, genetically engineered cell product is under investigation in a Phase 1 clinical trial (NCT03215004). Trial participants were conditioned with cyclophosphamide approximately 1 week before receiving a one-time low (< 109 genetically modified CD4+ T cells) or high (≥109 genetically modified CD4+ T cells) dose of AGT103-T, delivering between 2 and 21 million genetically modified cells per kilogram (kg) body weight. There were no serious adverse events (SAEs) and all adverse events (AEs) were mild. Genetically modified AGT103-T cells were detected in most of the participant blood samples collected 6 months after infusion, which was the last scheduled monitoring visit. Peripheral blood mononuclear cells (PBMC) collected after cell product infusion were tested to determine the abundance of Gag-specific T cells as a measure of objective responses to therapy. Gag-specific CD4+ T cells were detected in all treated individuals and were substantially increased by 9 to 300-fold compared to baseline, by 14 days after cell product infusion. Gag-specific CD8+ T cells were increased by 1.7 to 10-fold relative to baseline, by 28 days after cell product infusion. Levels of Gag-specific CD4+ T cells remained high (~2 to 70-fold higher relative to baseline) throughout 3-6 months after infusion. AGT103-T at low or high doses was safe and effective for improving host T cell immunity to HIV. Further studies, including antiretroviral treatment interruption, are warranted to evaluate the product's efficacy in HIV disease. Clinical trial registration: www.clinicaltrials.gov, identifier: NCT03215004.

18.
Cancer Immunol Immunother ; 60(3): 361-70, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21107834

RESUMEN

Clinical strategies to exploit Vγ2Vδ2 T cell responses for immunotherapy are confronted with short-term increases in cell levels or activity and the development of anergy that reduces the response to therapy with succeeding treatments. We are exploring strategies to increase the yield and durability of elicited Vγ2Vδ2 T cell responses. One approach focuses on the mammalian target of rapamycin (mTOR), which is important for regulating T cell metabolism and function. In Vγ2Vδ2 T cells, mTOR phosphorylates the S6K1 and eIF4EBP1 signaling intermediates after antigen stimulation. Rapamycin inhibited these phosphorylation events without impacting Akt or Erk activation, even though specific inhibition of Akt or Erk in turn reduced the activation of mTOR. The effects of rapamycin on the T cell receptor signaling pathway lead to increased proliferation of treated and antigen-exposed Vγ2Vδ2 cells. Rapamycin altered the phenotype of antigen-specific Vγ2Vδ2 cells by inducing a population shift from CD62L + CD69- to CD62L-CD69+, higher expression of CD25 or Bcl-2, lower levels of CCR5 and increased resistance to Fas-mediated cellular apoptosis. These changes were consistent with rapamycin promoting cell activation while decreasing the susceptibility to cell death that might occur by CCR5 or Fas signaling. Rapamycin treatment during antigen-stimulation of Vγ2Vδ2 T cells may be a strategy for overcoming current obstacles in tumor immunotherapy.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Sirolimus/farmacología , Linfocitos T/efectos de los fármacos , Células Presentadoras de Antígenos/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Técnicas In Vitro , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Activación de Linfocitos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores CCR5/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
19.
Blood ; 114(6): 1196-204, 2009 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-19498020

RESUMEN

Immune deficiency viruses such as SIV in macaques or HIV-1 in human beings have evolved mechanisms to defeat host immunity that also impact the efficacy of vaccines. A key factor for vaccine protection is whether immune responses elicited by prior immunization remain at levels sufficient to limit disease progression once a host is exposed to the pathogen. One potential mechanism for escaping pre-existing immunity is to trigger death among antigen-activated cells. We tested whether FasL/CD178 is involved in destroying preexisting immunity. Rhesus macaques were immunized with recombinant vesicular stomatitis virus vaccine expressing SIV Gag to elicit cellular immune responses, then treated with antibody that neutralizes FasL and challenged with intravenous SIVmac251. Compared with animals injected with control antibody, anti-FasL-treated macaques had superior preservation of central memory CD4(+) and CD8(+) cells and decreased regulatory T cells in the blood. The CD4(+) and CD8(+) lymphocytes from treated animals responded better to SIV Gag compared with controls, evidenced by higher cell-mediated immune responses to viral antigens for at least 17 weeks after SIV challenge. Anti-FasL treatment during the initial stages of acute SIV infection preserved the T-cell compartment and sustained cell-mediated immunity to SIV.


Asunto(s)
Anticuerpos/farmacología , Proteína Ligando Fas/inmunología , Productos del Gen gag/inmunología , Memoria Inmunológica , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Proteína Ligando Fas/antagonistas & inhibidores , VIH-1/inmunología , Humanos , Inmunidad Celular , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/terapia , Linfocitos T Reguladores/inmunología
20.
Curr Opin HIV AIDS ; 16(2): 83-87, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33625039

RESUMEN

PURPOSE OF REVIEW: Tremendous advances in cell and gene therapy may soon realize the goal of treating and possibly curing HIV disease. These advances rely on new technologies for cell engineering and new strategies for product manufacturing that are targeting the most important immune deficits in HIV and promising to reconstitute protective, antiviral immunity and achieve natural suppression of HIV disease. RECENT FINDINGS: We summarize important advances in vectored passive immunity, e.g., directing in vivo expression of protective antibodies or antiviral proteins, B cell engineering to overcome the inadequate humoral immune response to HIV, and T cell engineering that is breaking new ground using viral vector modification of HIV specific T cells. These innovative approaches build on a substantial history of gene and cell therapy research in HIV disease. SUMMARY: Cell and gene therapy for HIV disease has been an area of tremendous innovation during the nearly two decades since early reports showed evidence for modulating disease. Recent efforts are building on the early experiences, closing gaps in previous approaches, and moving closer to effective treatment. Products approaching or already in clinical trials hold great promise for achieving durable suppression of HIV that will revolutionize therapy and offering hope to infected individuals that disease may be controlled without lifelong dependence on antiretroviral medications. VIDEO ABSTRACT: http://links.lww.com/COHA/A15.


Asunto(s)
Terapia Genética , Infecciones por VIH , Antirretrovirales/uso terapéutico , Antivirales/uso terapéutico , Vectores Genéticos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/terapia , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA