Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 201(1): 124-133, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29752313

RESUMEN

Epigenetic mechanisms, such as DNA methylation, determine immune cell phenotype. To understand the epigenetic alterations induced by helminth coinfections, we evaluated the longitudinal effect of ascariasis and schistosomiasis infection on CD4+ T cell DNA methylation and the downstream tuberculosis (TB)-specific and bacillus Calmette-Guérin-induced immune phenotype. All experiments were performed on human primary immune cells from a longitudinal cohort of recently TB-exposed children. Compared with age-matched uninfected controls, children with active Schistosoma haematobium and Ascaris lumbricoides infection had 751 differentially DNA-methylated genes, with 72% hypermethylated. Gene ontology pathway analysis identified inhibition of IFN-γ signaling, cellular proliferation, and the Th1 pathway. Targeted real-time quantitative PCR after methyl-specific endonuclease digestion confirmed DNA hypermethylation of the transcription factors BATF3, ID2, STAT5A, IRF5, PPARg, RUNX2, IRF4, and NFATC1 and cytokines or cytokine receptors IFNGR1, TNFS11, RELT (TNF receptor), IL12RB2, and IL12B (p < 0.001; Sidak-Bonferroni). Functional blockage of the IFN-γ signaling pathway was confirmed, with helminth-infected individuals having decreased upregulation of IFN-γ-inducible genes (Mann-Whitney p < 0.05). Hypomethylation of the IL-4 pathway and DNA hypermethylation of the Th1 pathway was confirmed by Ag-specific multidimensional flow cytometry demonstrating decreased TB-specific IFN-γ and TNF and increased IL-4 production by CD4+ T cells (Wilcoxon signed-rank p < 0.05). In S. haematobium-infected individuals, these DNA methylation and immune phenotypic changes persisted at least 6 mo after successful deworming. This work demonstrates that helminth infection induces DNA methylation and immune perturbations that inhibit TB-specific immune control and that the duration of these changes are helminth specific.


Asunto(s)
Ascariasis/inmunología , Ascaris lumbricoides/inmunología , Vacuna BCG/inmunología , Metilación de ADN/genética , Schistosoma haematobium/inmunología , Esquistosomiasis/inmunología , Células TH1/inmunología , Animales , Proliferación Celular/fisiología , Células Cultivadas , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-4/biosíntesis , Interleucina-4/genética , Receptores de Citocinas/genética , Factores de Transcripción/genética , Tuberculosis/inmunología
2.
PLoS Pathog ; 12(8): e1005805, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27486665

RESUMEN

The loss of HIV-specific CD8+ T cell cytolytic function is a primary factor underlying progressive HIV infection, but whether HIV-specific CD8+ T cells initially possess cytolytic effector capacity, and when and why this may be lost during infection, is unclear. Here, we assessed CD8+ T cell functional evolution from primary to chronic HIV infection. We observed a profound expansion of perforin+ CD8+ T cells immediately following HIV infection that quickly waned after acute viremia resolution. Selective expression of the effector-associated transcription factors T-bet and eomesodermin in cytokine-producing HIV-specific CD8+ T cells differentiated HIV-specific from bulk memory CD8+ T cell effector expansion. As infection progressed expression of perforin was maintained in HIV-specific CD8+ T cells with high levels of T-bet, but not necessarily in the population of T-betLo HIV-specific CD8+ T cells that expand as infection progresses. Together, these data demonstrate that while HIV-specific CD8+ T cells in acute HIV infection initially possess cytolytic potential, progressive transcriptional dysregulation leads to the reduced CD8+ T cell perforin expression characteristic of chronic HIV infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Infecciones por VIH/inmunología , Inmunidad Celular , Adulto , Linfocitos T CD8-positivos/patología , Enfermedad Crónica , Femenino , Infecciones por VIH/patología , Humanos , Masculino , Persona de Mediana Edad , Perforina/inmunología , Proteínas de Dominio T Box/inmunología
3.
Blood ; 128(26): 3061-3072, 2016 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-27799162

RESUMEN

Autosomal dominant hyper-IgE syndrome (AD-HIES) is caused by dominant-negative mutations in STAT3; however, the molecular basis for mutant STAT3 allele dysfunction is unclear and treatment remains supportive. We hypothesized that AD-HIES mutations decrease STAT3 protein stability and that mutant STAT3 activity can be improved by agents that increase chaperone protein activity. We used computer modeling to characterize the effect of STAT3 mutations on protein stability. We measured STAT3 protein half-life (t1/2) and determined levels of STAT3 phosphorylated on tyrosine (Y) 705 (pY-STAT3) and mRNA levels of STAT3 gene targets in Epstein-Barr virus-transformed B (EBV) cells, human peripheral blood mononuclear cells (PBMCs), and mouse splenocytes incubated without or with chaperone protein modulators-HSF1A, a small-molecule TRiC modulator, or geranylgeranylacetone (GGA), a drug that upregulates heat shock protein (HSP) 70 and HSP90. Computer modeling predicted that 81% of AD-HIES mutations are destabilizing. STAT3 protein t1/2 in EBV cells from AD-HIES patients with destabilizing STAT3 mutations was markedly reduced. Treatment of EBV cells containing destabilizing STAT3 mutations with either HSF1A or GGA normalized STAT3 t1/2, increased pY-STAT3 levels, and increased mRNA levels of STAT3 target genes up to 79% of control. In addition, treatment of human PBMCs or mouse splenocytes containing destabilizing STAT3 mutations with either HSF1A or GGA increased levels of cytokine-activated pY-STAT3 within human CD4+ and CD8+ T cells and numbers of IL-17-producing CD4+ mouse splenocytes, respectively. Thus, most AD-HIES STAT3 mutations are destabilizing; agents that modulate chaperone protein function improve STAT3 stability and activity in T cells and may provide a specific treatment.


Asunto(s)
Síndrome de Job/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Citocinas/farmacología , Proteínas de Unión al ADN/metabolismo , Diterpenos/farmacología , Semivida , Factores de Transcripción del Choque Térmico , Herpesvirus Humano 4/fisiología , Humanos , Interleucina-17/metabolismo , Síndrome de Job/patología , Ratones , Modelos Moleculares , Proteínas Mutantes/metabolismo , Mutación/genética , Fosfotirosina/metabolismo , Unión Proteica/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Factor de Transcripción STAT3/química , Factor de Transcripción STAT3/genética , Bazo/patología , Factores de Transcripción/metabolismo
4.
Blood ; 125(4): 591-9, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25359994

RESUMEN

Germline loss-of-function mutations in the transcription factor signal transducer and activator of transcription 3 (STAT3) cause immunodeficiency, whereas somatic gain-of-function mutations in STAT3 are associated with large granular lymphocytic leukemic, myelodysplastic syndrome, and aplastic anemia. Recently, germline mutations in STAT3 have also been associated with autoimmune disease. Here, we report on 13 individuals from 10 families with lymphoproliferation and early-onset solid-organ autoimmunity associated with 9 different germline heterozygous mutations in STAT3. Patients exhibited a variety of clinical features, with most having lymphadenopathy, autoimmune cytopenias, multiorgan autoimmunity (lung, gastrointestinal, hepatic, and/or endocrine dysfunction), infections, and short stature. Functional analyses demonstrate that these mutations confer a gain-of-function in STAT3 leading to secondary defects in STAT5 and STAT1 phosphorylation and the regulatory T-cell compartment. Treatment targeting a cytokine pathway that signals through STAT3 led to clinical improvement in 1 patient, suggesting a potential therapeutic option for such patients. These results suggest that there is a broad range of autoimmunity caused by germline STAT3 gain-of-function mutations, and that hematologic autoimmunity is a major component of this newly described disorder. Some patients for this study were enrolled in a trial registered at www.clinicaltrials.gov as #NCT00001350.


Asunto(s)
Enfermedades Autoinmunes/genética , Enfermedades Genéticas Congénitas/genética , Trastornos Linfoproliferativos/genética , Factor de Transcripción STAT3/genética , Adolescente , Adulto , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Niño , Preescolar , Femenino , Enfermedades Genéticas Congénitas/inmunología , Enfermedades Genéticas Congénitas/patología , Humanos , Lactante , Trastornos Linfoproliferativos/inmunología , Trastornos Linfoproliferativos/patología , Masculino , Mutación , Fosforilación/genética , Fosforilación/inmunología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología
5.
J Infect Dis ; 214(3): 479-88, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27389351

RESUMEN

Helminth-infected individuals possess a higher risk of developing tuberculosis, but the precise immunologic mechanism of Mycobacterium tuberculosis control remains unclear. We hypothesized that a perturbation of the M. tuberculosis-specific CD4(+) T-cell response weakens the ability of macrophages to contain M. tuberculosis We exposed peripheral blood mononuclear cells from M. tuberculosis-infected humans to schistosome soluble egg antigen (SEA) and then profiled M. tuberculosis-specific CD4(+) T cells via multiparametric flow cytometry. SEA decreased the frequency of cells producing interferon γ (6.79% vs 3.20%; P = .017) and tumor necrosis factor α (6.98% vs 2.96%; P = .012), with a concomitant increase in the median fluorescence intensity of interleukin 4 (IL-4; P < .05) and interleukin 10 (IL-10; 1440 vs 1273; P < .05). Macrophages polarized with SEA-exposed, autologous CD4(+) T-cell supernatant had a 2.19-fold decreased colocalization of lysosomes and M. tuberculosis (P < .05). When polarized with IL-4 or IL-10, macrophages had increased expression of CD206 (P < .0001), 1.5-fold and 1.9 fold increased intracellular numbers of M. tuberculosis per macrophage (P < .0005), and 1.4-fold and 1.7-fold decreased colocalization between M. tuberculosis and lysosomes (P < .001). This clarifies a relationship in which helminth-induced CD4(+) T cells disrupt M. tuberculosis control by macrophages, thereby providing a mechanism for the observation that helminth infection advances the progression of tuberculosis among patients with M. tuberculosis infection.


Asunto(s)
Antígenos Helmínticos/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Factores Inmunológicos/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Mycobacterium tuberculosis/inmunología , Schistosoma/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Citocinas/biosíntesis , Citometría de Flujo , Humanos , Tolerancia Inmunológica , Inmunofenotipificación , Leucocitos Mononucleares/inmunología , Lisosomas/metabolismo , Macrófagos/fisiología , Fagosomas/metabolismo
6.
J Infect Dis ; 213(11): 1701-7, 2016 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26908740

RESUMEN

The ability to distinguish allogeneic hematopoietic cell transplant (allo-HCT) recipients at risk for cytomegalovirus (CMV) reactivation from those who are not is central for optimal CMV management strategies. Interferon γ (IFN-γ) produced by CMV-challenged T cells may serve as an immune marker differentiating these 2 populations. We prospectively monitored 63 CMV-seropositive allo-HCT recipients with a CMV-specific enzyme-linked immunospot (ELISPOT) assay and for CMV infection from the period before transplantation to day 100 after transplantation. Assay results above certain thresholds (50 spots per 250 000 cells for immediate early 1 or 100 spots per 250 000 cells for phosphoprotein 65) identified patients who were protected against CMV infection as long as they had no graft-versus-host disease and/or were not receiving systemic corticosteroids. Based on the multivariable Cox proportional hazards regression model, the only significant factor for preventing CMV reactivation was a CMV-specific ELISPOT response above the determined thresholds (adjusted hazard ratio, 0.21; 95% confidence interval, .05-.97; P = .046). Use of this assay as an additional tool for managing allo-HCT recipients at risk for CMV reactivation needs further validation in future studies. Application of this new approach may reduce the duration and intensity of CMV monitoring and the duration of prophylaxis or treatment with antiviral agents in those who have achieved CMV-specific immune reconstitution.


Asunto(s)
Infecciones por Citomegalovirus/diagnóstico , Ensayo de Immunospot Ligado a Enzimas , Trasplante de Células Madre Hematopoyéticas , Ensayos de Liberación de Interferón gamma , Receptores de Trasplantes , Adulto , Anciano , Biomarcadores/sangre , Estudios de Cohortes , Infecciones por Citomegalovirus/complicaciones , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Interferón gamma/sangre , Leucemia/complicaciones , Leucemia/terapia , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Medición de Riesgo , Linfocitos T/inmunología , Adulto Joven
7.
Mediators Inflamm ; 2016: 1478340, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27006526

RESUMEN

INTRODUCTION: Our objective is to understand how HIV infection increases the risk of progression from latent tuberculosis (TB) to active disease. We understand now that immunity is a balance of competing immune responses by multiple cell types. Since T-lymphocyte production of interferon-gamma (IFN-γ) in response to Mycobacterium tuberculosis (Mtb) antigens fails to differentiate disease from latent infection, we applied a comprehensive profiling methodology to define immune biomarkers that reliably predict a patient's TB risk. METHODS: We established a cohort of HIV-infected adults with TB disease from Swaziland. Multiparametric flow cytometry was used to quantify the mycobacterial-specific anti-inflammatory (IL-4 and IL-10) and proinflammatory (IFN-γ) immune response. RESULTS: From 12 HIV-infected Swaziland patients with TB disease, the CD4(+), CD8(+), Double Negative, and CD56(+)CD3(-) lymphocytes increase their IL-4 : IFN-γ ratio as HIV disease worsens (Spearman r of -0.59; -0.59; -0.60; and -0.59, resp.; p < 0.05). Similarly, HIV severity is associated with an increased IL-10 : IFN-γ ratio (Spearman r of -0.76; p = 0.01). CONCLUSION: As HIV disease progresses, both the adaptive and innate branches skew away from an inflammatory and towards anti-inflammatory phenotype.


Asunto(s)
Inmunidad Adaptativa/fisiología , Infecciones por VIH/complicaciones , Infecciones por VIH/inmunología , Inmunidad Innata/fisiología , Mycobacterium tuberculosis/inmunología , Inmunidad Adaptativa/genética , Adolescente , Adulto , Antígenos Bacterianos/inmunología , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Antígeno CD56/metabolismo , Linfocitos T CD8-positivos/metabolismo , Femenino , Humanos , Inmunidad Innata/genética , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Masculino , Mycobacterium tuberculosis/patogenicidad , Adulto Joven
8.
J Allergy Clin Immunol ; 135(5): 1293-302, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25748067

RESUMEN

BACKGROUND: DiGeorge syndrome affects more than 3.5 million persons worldwide. Partial DiGeorge syndrome (pDGS), which is characterized by a number of gene deletions in chromosome 22, including the chicken tumor virus number 10 regulator of kinase (Crk)-like (CrkL) gene, is one of the most common genetic disorders in human subjects. To date, the role of natural killer (NK) cells in patients with pDGS remains unclear. OBJECTIVE: We sought to define the effect of pDGS-related Crk haploinsufficiency on NK cell activation and cytotoxic immunological synapse (IS) structure and function. METHODS: Inducible CrkL-silenced NK cells were used to recapitulate the pDGS, CrkL-haploinsufficient phenotype. Findings were validated by using NK cells from patients with actual pDGS. Ultimately, deficits in the function of NK cells from patients with pDGS were restored by lentiviral transduction of CrkL. RESULTS: Silencing of CrkL expression inhibits NK cell function. Specifically, pDGS haploinsufficiency of CrkL inhibits accumulation of activating receptors, polarization of cytolytic machinery and key signaling molecules, and activation of ß2-integrin at the IS. Reintroduction of CrkL protein restores NK cell cytotoxicity. CONCLUSION: CrkL haploinsufficiency causes functional NK deficits in patients with pDGS by disrupting both ß2-integrin activation and activating receptor accumulation at the IS. Our results suggest that NK cell IS quality can directly affect immune status, providing a potential target for diagnosis and therapeutic manipulation in patients with pDGS and in other patients with functional NK cell deficiencies.


Asunto(s)
Síndrome de DiGeorge/genética , Síndrome de DiGeorge/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Citotoxicidad Inmunológica/genética , Silenciador del Gen , Humanos , Sinapsis Inmunológicas/genética , Sinapsis Inmunológicas/metabolismo , Integrinas/metabolismo , Modelos Biológicos , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo
9.
Blood ; 121(14): 2669-77, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23365458

RESUMEN

Mutations in the transcription factor GATA2 underlie the syndrome of monocytopenia and B- and natural killer (NK)-cell lymphopenia associated with opportunistic infections and cancers. In addition, patients have recurrent and severe viral infections. NK cells play a critical role in mediating antiviral immunity. Human NK cells are thought to mature in a linear fashion, with the CD56(bright) stage preceding terminal maturation to the CD56(dim) stage, considered the most enabled for cytotoxicity. Here we report an NK cell functional defect in GATA2-deficient patients and extend this genetic lesion to what is considered to be the original NK cell-deficient patient. In most cases, GATA2 deficiency is accompanied by a severe reduction in peripheral blood NK cells and marked functional impairment. The NK cells detected in peripheral blood of some GATA2-deficient patients are exclusively of the CD56(dim) subset, which is recapitulated on in vitro NK cell differentiation. In vivo, interferon α treatment increased NK cell number and partially restored function but did not correct the paucity of CD56(bright) cells. Thus, GATA2 is required for the maturation of human NK cells and the maintenance of the CD56(bright) pool in the periphery. Defects in GATA2 are a novel cause of profound NK cell dysfunction.


Asunto(s)
Antígeno CD56/inmunología , Diferenciación Celular/inmunología , Factor de Transcripción GATA2/genética , Células Asesinas Naturales/inmunología , Linfopenia/genética , Antígenos CD34/metabolismo , Antígeno CD56/metabolismo , Citotoxicidad Inmunológica/inmunología , Factor de Transcripción GATA2/inmunología , Factor de Transcripción GATA2/metabolismo , Humanos , Inmunofenotipificación , Células K562 , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/inmunología , Recuento de Linfocitos , Linfopenia/inmunología , Linfopenia/metabolismo , Células del Estroma/citología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
10.
J Immunol ; 190(7): 3207-15, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23455505

RESUMEN

In mice, two T-box transcription factors, T-box expressed in T cells (T-bet) and eomesodermin (Eomes), drive the differentiation of CD8 T cell lineages; however, little is known regarding their role in human CD8 T cell differentiation. In this study, we characterized T-bet and Eomes expression and localization within human CD8 memory T cell populations. We find that T-bet and Eomes are broadly expressed in human memory CD8 T cells, with increasing levels of T-bet and Eomes strongly correlating with differentiation from central memory to effector memory and effector subpopulations. In resting T cells, T-bet levels directly correlate to subcellular localization, with a higher propensity for nuclear expression of T-bet within T-bet(hi) cells and predominantly cytoplasmic expression in T-bet(lo) cells. In addition, Eomes is also localized to either the nucleus or the cytoplasm. Upon TCR stimulation, the percentage of T cells that express T-bet dramatically increases, whereas the percentage of cells expressing Eomes remains largely unchanged across all memory populations. Of interest, T-bet, but not Eomes, relocalizes to the nucleus in the majority of cells across all populations within 24 h post stimulation. These data indicate that T-bet and Eomes are likely regulated at the level of subcellular localization, potentially via different mechanisms. Together, these findings suggest a novel model for CD8 T cell differentiation in humans that is based on the localization of T-bet and Eomes.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Memoria Inmunológica , Proteínas de Dominio T Box/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Humanos , Inmunofenotipificación , Activación de Linfocitos/inmunología , Fenotipo , Transporte de Proteínas , Receptores de Antígenos de Linfocitos T/metabolismo
11.
Immunol Rev ; 239(1): 109-24, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21198668

RESUMEN

The Merck STEP and the Thai RV144 human immunodeficiency virus (HIV) vaccine trials confirmed that we still have a long way to go before developing a prophylactic HIV vaccine. The main issue at hand is that we have yet to identify an immunological correlate of protection against HIV. While many question the T-cell-based approach towards vaccine development, it is likely that T cells will be a necessary part of any vaccine strategy. CD8(+) T cells remain an attractive option because of their ability to specifically recognize and eliminate virally infected host cells. In this review, we recapitulate the evidence for CD8(+) T cells as an immunological correlate against HIV, but more importantly, we assess the means by which we evaluate their antiviral capacity. To achieve a breakthrough in the domain of T-cell-based HIV vaccine development, it has become abundantly clear that we must overhaul our system of immune monitoring and come up with a 'rational' tactic to evaluate the efficacy of HIV-specific CD8(+) T cells.


Asunto(s)
Vacunas contra el SIDA/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/prevención & control , VIH/inmunología , Vacunas contra el SIDA/genética , Inmunidad Adaptativa , Ensayos Clínicos como Asunto , Citocinas/inmunología , Citocinas/metabolismo , VIH/fisiología , Infecciones por VIH/inmunología , Humanos , Activación de Linfocitos , Perforina/metabolismo
12.
J Clin Immunol ; 33(7): 1223-39, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23888327

RESUMEN

PURPOSE & METHODS: The immunopathogenic mechanisms responsible for debilitating neurodegenerative and oncologic diseases associated with human T-cell leukemia virus type 1 (HTLV-1) are not fully understood. Quality of cytotoxic T lymphocytes (CTLs) is being increasingly associated with the outcome of persistent HTLV-1 infection. In this respect, a patient cohort (from HTLV-1 endemic region) consisting of seronegative controls (controls), asymptomatic carriers (ACs), and patients with adult T-cell leukemia (ATL) or HTLV-associated myelopathy/tropical spastic paraparesis (HAM/TSP) was analyzed for CD8(+) T cells polyfunctionality in response to the viral antigen Tax. RESULTS: Compared to ACs, ATL and HAM/TSP patients had lower frequency and polyfunctionality of CTLs in response to Tax suggesting dysfunction of CD8(+) T cells in these individuals. As an underlying mechanism, programmed death-1 (PD-1) receptor was found to be highly unregulated in Tax-responsive as well as total CD8(+) T cells from ATL and HAM/TSP but not from ACs and directly correlated with the lack of polyfunctionality in these individuals. Further, PD-1 expression showed a direct whereas MIP-1α expression had an indirect correlation with the proviral load providing new insights about the immunopathogenesis of HTLV-associated diseases. Additionally, we identified key cytokine signatures defining the immune activation status of clinical samples by the luminex assay. CONCLUSIONS: Collectively, our findings suggest that reconstitution of fully functional CTLs, stimulation of MIP-1α expression, and/or blockade of the PD-1 pathway are potential approaches for immunotherapy / therapeutic vaccine against HTLV-mediated diseases.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Virus Linfotrópico T Tipo 1 Humano/inmunología , Leucemia-Linfoma de Células T del Adulto/inmunología , Paraparesia Espástica Tropical/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Adolescente , Adulto , Anciano , Enfermedades Asintomáticas , Linfocitos T CD8-positivos/virología , Quimiocina CCL3/genética , Quimiocina CCL3/metabolismo , Citocinas/genética , Citocinas/metabolismo , Citotoxicidad Inmunológica , Femenino , Regulación de la Expresión Génica , Productos del Gen tax/inmunología , Virus Linfotrópico T Tipo 1 Humano/crecimiento & desarrollo , Humanos , Activación de Linfocitos , Recuento de Linfocitos , Persona de Mediana Edad , Receptor de Muerte Celular Programada 1/genética , Transcriptoma , Carga Viral , Adulto Joven
13.
Blood ; 117(14): 3799-808, 2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-21289310

RESUMEN

Recent data suggest that CD8+ T-cell effector activity is an important component in the control of HIV replication in elite controllers (ECs). One critical element of CD8+ T-cell effector function and differentiation is the T-box transcription factor T-bet. In the present study, we assessed T-bet expression, together with the effector proteins perforin, granzyme A (Grz A), granzyme B (Grz B), and granulysin, in HIV-specific CD8+ T cells from ECs (n = 20), chronically infected progressors (CPs; n = 18), and highly active antiretroviral therapy (HAART)-suppressed individuals (n = 19). Compared with the other cohort groups, HIV-specific CD8+ T cells among ECs demonstrated a superior ability to express perforin and Grz B, but with no detectable difference in the levels of Grz A or granulysin. We also observed higher levels of T-bet in HIV-specific CD8+ T cells from ECs, with an ensuing positive correlation between T-bet and levels of both perforin and Grz B. Moreover, HIV-specific CD8+ T cells in ECs up-regulated T-bet to a greater extent than CPs after in vitro expansion, with concomitant up-regulation of perforin and Grz B. These results suggest that T-bet may play an important role in driving effector function, and its modulation may lead to enhanced effector activity against HIV.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Infecciones por VIH/inmunología , VIH/inmunología , Proteínas de Dominio T Box/metabolismo , Fármacos Anti-VIH/uso terapéutico , Terapia Antirretroviral Altamente Activa , Linfocitos T CD8-positivos/patología , Estudios de Cohortes , Estudios Transversales , Progresión de la Enfermedad , Granzimas/metabolismo , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/metabolismo , Infecciones por VIH/patología , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/inmunología , Tolerancia Inmunológica/fisiología , Perforina/metabolismo , Proteínas de Dominio T Box/fisiología , Especificidad del Receptor de Antígeno de Linfocitos T , Resultado del Tratamiento
14.
PLoS Pathog ; 6(3): e1000798, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20221423

RESUMEN

The prevailing paradigm of T lymphocyte control of viral replication is that the protective capacity of virus-specific CD8(+) T cells is directly proportional to the number of functions they can perform, with IL-2 production capacity considered critical. Having recently defined rapid perforin upregulation as a novel effector function of antigen-specific CD8(+) T cells, here we sought to determine whether new perforin production is a component of polyfunctional CD8(+) T cell responses that contributes to the control of several human viral infections: cytomegalovirus (CMV), Epstein-Barr virus (EBV), influenza (flu), and adenovirus (Ad). We stimulated normal human donor PBMC with synthetic peptides whose amino acid sequences correspond to defined CTL epitopes in the aforementioned viruses, and then used polychromatic flow cytometry to measure the functional capacity and the phenotype of the responding CD8(+) T cells. While EBV and flu-specific CD8(+) T cells rarely upregulate perforin, CMV-specific cells often do and Ad stimulates an exceptionally strong perforin response. The differential propensity of CD8(+) T cells to produce either IL-2 or perforin is in part related to levels of CD28 and the transcription factor T-bet, as CD8(+) T cells that rapidly upregulate perforin harbor high levels of T-bet and those producing IL-2 express high amounts of CD28. Thus, "polyfunctional" profiling of antigen-specific CD8(+) T cells must not be limited to simply the number of functions the cell can perform, or one particular memory phenotype, but should actually define which combinations of memory markers and functions are relevant in each pathogenic context.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Interleucina-2/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Virosis/inmunología , Antígenos CD28/inmunología , Antígenos CD28/metabolismo , Linfocitos T CD8-positivos/metabolismo , Infecciones por Citomegalovirus/inmunología , Infecciones por Virus de Epstein-Barr/inmunología , Humanos , Memoria Inmunológica/inmunología , Inmunofenotipificación , Gripe Humana/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-2/inmunología , Activación de Linfocitos/inmunología , Perforina , Proteínas Citotóxicas Formadoras de Poros/inmunología , Proteínas de Dominio T Box/inmunología , Proteínas de Dominio T Box/metabolismo , Regulación hacia Arriba/inmunología
15.
PLoS Pathog ; 6(5): e1000917, 2010 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-20523897

RESUMEN

Many immune correlates of CD8(+) T-cell-mediated control of HIV replication, including polyfunctionality, proliferative ability, and inhibitory receptor expression, have been discovered. However, no functional correlates using ex vivo cells have been identified with the known ability to cause the direct elimination of HIV-infected cells. We have recently discovered the ability of human CD8(+) T-cells to rapidly upregulate perforin--an essential molecule for cell-mediated cytotoxicity--following antigen-specific stimulation. Here, we examined perforin expression capability in a large cross-sectional cohort of chronically HIV-infected individuals with varying levels of viral load: elite controllers (n = 35), viremic controllers (n = 29), chronic progressors (n = 27), and viremic nonprogressors (n = 6). Using polychromatic flow cytometry and standard intracellular cytokine staining assays, we measured perforin upregulation, cytokine production, and degranulation following stimulation with overlapping peptide pools encompassing all proteins of HIV. We observed that HIV-specific CD8(+) T-cells from elite controllers consistently display an enhanced ability to express perforin directly ex vivo compared to all other groups. This ability is not restricted to protective HLA-B haplotypes, does not require proliferation or the addition of exogenous factors, is not restored by HAART, and primarily originates from effector CD8(+) T-cells with otherwise limited functional capability. Notably, we found an inverse relationship between HIV-specific perforin expression and viral load. Thus, the capability of HIV-specific CD8(+) T-cells to rapidly express perforin defines a novel correlate of control in HIV infection.


Asunto(s)
Linfocitos T CD8-positivos , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Vacunas contra el SIDA/inmunología , Inmunidad Adaptativa/genética , Inmunidad Adaptativa/inmunología , Alelos , Terapia Antirretroviral Altamente Activa , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Estudios de Cohortes , Estudios Transversales , Citocinas/metabolismo , Progresión de la Enfermedad , Citometría de Flujo , Infecciones por VIH/tratamiento farmacológico , Antígenos HLA-B/genética , Haplotipos , Humanos , Perforina , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/virología , Carga Viral/inmunología , Replicación Viral/inmunología
16.
J Immunol ; 182(9): 5560-9, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19380804

RESUMEN

CTL are endowed with the ability to eliminate pathogens through perforin-mediated cytotoxic activity. The mechanism for perforin-mediated Ag-specific killing has been solely attributed to cytotoxic granule exocytosis from activated CD8(+) T cells. In this study, we redefine this mechanism, demonstrating that virus-specific CD8(+) T cells rapidly up-regulate perforin in response to stimulation temporally with IFN-gamma and CD107a expression. Following Ag-specific activation, newly synthesized perforin rapidly appears at the immunological synapse, both in association with and independent of cytotoxic granules, where it functions to promote cytotoxicity. Our work suggests a novel mechanism of CTL cytotoxicity and identifies a novel correlate of CD8(+) T cell-mediated immunity.


Asunto(s)
Gránulos Citoplasmáticos/inmunología , Pruebas Inmunológicas de Citotoxicidad , Epítopos de Linfocito T/inmunología , Sinapsis Inmunológicas/inmunología , Perforina/biosíntesis , Perforina/fisiología , Linfocitos T Citotóxicos/inmunología , Regulación hacia Arriba/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Degranulación de la Célula/inmunología , Línea Celular Transformada , Gránulos Citoplasmáticos/metabolismo , Pruebas Inmunológicas de Citotoxicidad/métodos , Humanos , Activación de Linfocitos/inmunología , Perforina/metabolismo , Transporte de Proteínas/inmunología , Transducción de Señal/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/virología
17.
J Immunol ; 183(1): 706-17, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19542473

RESUMEN

Our limited understanding of the interaction between primate lentiviruses and the host immune system complicates the design of an effective HIV/AIDS vaccine. To identify immunological correlates of protection from SIV disease progression, we immunized two groups of five rhesus macaques (RMs) with either modified vaccinia Ankara (MVA) or MVADeltaudg vectors that expressed SIVmac239 Gag and Tat. Both vectors raised a SIV-specific CD8(+) T cell response, with a magnitude that was greater in mucosal tissues than in peripheral blood. After challenge with SIVmac239, all vaccinated RMs showed mucosal and systemic CD8(+) T cell recall responses that appeared faster and were of greater magnitude than those in five unvaccinated control animals. All vaccinated RMs showed a approximately 1-log lower peak and early set-point SIV viral load than the unvaccinated animals, and then, by 8 wk postchallenge, exhibited levels of viremia similar to the controls. We observed a significant direct correlation between the magnitude of postchallenge SIV-specific CD8(+) T cell responses and SIV viral load. However, vaccinated RMs showed no protection from either systemic or mucosal CD4(+) T cell depletion and no improved survival. The observation that vaccine-induced, SIV-specific CD8(+) T cells that partially control SIVmac239 virus replication fail to protect from immunological or clinical progression of SIV infection underscores both the complexity of AIDS pathogenesis and the challenges of properly assessing the efficacy of candidate AIDS vaccines.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Progresión de la Enfermedad , Epítopos de Linfocito T/inmunología , Activación de Linfocitos/inmunología , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Replicación Viral/inmunología , Animales , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/virología , Macaca mulatta , Vacunas contra el SIDAS/administración & dosificación , Vacunas contra el SIDAS/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control
18.
J Immunol Methods ; 492: 112998, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33600819

RESUMEN

An integrated understanding of the functional capacities of cells in the context of their physical parameters and molecular markers is increasingly demanded in immunologic studies. Regulatory T cells (Tregs) are a subpopulation of T cells involved in immune response modulation and mediating tolerance to self-antigen with their absence leading to a loss of tolerance. Glycoprotein repetitions A predominant (GARP) is a key marker for activated Tregs, but its detection may also be useful in determining the functional capacities of the cell. This study aims to deduce the optimal stimulation period and the impact of protein transport inhibitors (PTIs), commonly used in the detection of intracellular cytokines, on GARP detection. Through flow cytometric analysis we analyzed different cell culture conditions for optimal GARP expression on activated Tregs. Healthy donor PBMCs were stimulated with either Staphylococcal Enterotoxin B (SEB) or PMA/Ionomycin (PMA/Iono), in the presence and absence of PTIs monensin and/or brefeldin A (BFA) and GARP expression was assessed on CD4+ CD25+ FOXP3+ Tregs. The optimal stimulation period for the detection of GARP was highest at 24-h. Furthermore, we determined that GARP expression on Tregs is significantly reduced when cells are treated with the PTIs monensin and/or BFA following PMA/Iono stimulation. This effect was not seen following SEB stimulation. Therefore, due to the effects of PTIs, alternative methods should be considered when performing simultaneous analysis for cytokine expression and GARP expression on Tregs.


Asunto(s)
Citocinas/análisis , Inmunofenotipificación/métodos , Proteínas de la Membrana/análisis , Linfocitos T Reguladores/inmunología , Brefeldino A/farmacología , Células Cultivadas , Citocinas/metabolismo , Enterotoxinas/inmunología , Voluntarios Sanos , Humanos , Activación de Linfocitos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Monensina/farmacología , Cultivo Primario de Células , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo
19.
Life Sci Space Res (Amst) ; 31: 29-33, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34689947

RESUMEN

During long duration orbital space missions, astronauts experience immune system dysregulation, the persistent reactivation of latent herpesviruses, and some degree of clinical incidence. During planned NASA 'Artemis' deep space missions the stressors that cause this phenomenon will increase, while clinical care capability will likely be reduced. There is currently minimal clinical laboratory capability aboard the International Space Station (ISS). The ability to monitor the white blood cell count (WBC) and differential during spaceflight has been an unmet NASA medical requirement, primarily due to a lack of capable hardware. We performed ground and flight validation of a device designed to monitor WBC and differential within minutes from a fingerstick blood sample. This device is miniaturized, robust, and generally compatible with microgravity operations. Ground testing for spaceflight consisted of vibration tolerance, power/battery and interface requirements, electromagnetic interference (EMI), and basic evaluation of sample preparation and operations in the context of spaceflight constraints. The in-flight validation performed aboard the ISS by two astronauts included assessment of three levels of control solution (blood) samples as well as a real time analysis of a fingerstick blood sample by one of the crewmembers. Flight and ground testing of the same lot of control solutions yielded similar total WBC values. There was some select discrepancy between flight and ground data for the differential analysis. However, the data suggest that this issue is due to compromise of the control solutions as a result of storage length before flight operations, and not due to a microgravity-associated issue with instrument performance. This evaluation also yielded lessons learned regarding crewmember training for technique-sensitive small-volume biosample collection and handling in microgravity. The fingerstick analysis was successful and was the first real-time hematology assessment performed during spaceflight. This device may provide an in-mission monitoring capability for astronauts thereby assisting Flight Surgeons and the crew medical officer during both orbital and deep space missions.


Asunto(s)
Vuelo Espacial , Ingravidez , Astronautas , Humanos , Sistemas de Atención de Punto , Tecnología , Ingravidez/efectos adversos
20.
J Allergy Clin Immunol Pract ; 8(10): 3247-3250, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32971311

RESUMEN

NASA implements required medical tests and clinical monitoring to ensure the health and safety of its astronauts. These measures include a pre-launch quarantine to mitigate the risk of infectious diseases. During space missions, most astronauts experience perturbations to their immune system that manifest as a detectable secondary immunodeficiency. On return to Earth, after the stress of re-entry and landing, astronauts would be most vulnerable to infectious disease. In April 2020, a crew returned from International Space Station to NASA Johnson Space Center in Houston, Texas, during the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. Post-flight quarantine protocols (both crew and contacts) were enhanced to protect this crew from SARS-CoV-2. In addition, specific additional clinical monitoring was performed to determine post-flight immunocompetence. Given that coronavirus disease 2019 (COVID-19) prognosis is more severe for the immunocompromised, a countermeasures protocol for spaceflight suggested by an international team of scientists could benefit terrestrial patients with secondary immunodeficiency.


Asunto(s)
Astronautas , Infecciones por Coronavirus/prevención & control , Huésped Inmunocomprometido/inmunología , Pandemias/prevención & control , Neumonía Viral/prevención & control , Cuarentena/métodos , Vuelo Espacial , Estrés Fisiológico/inmunología , Betacoronavirus , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , COVID-19 , Infecciones por Coronavirus/inmunología , Suplementos Dietéticos , Terapia por Ejercicio , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Inmunoglobulina G/uso terapéutico , Interleucina-2/uso terapéutico , Política Organizacional , Neumonía Viral/inmunología , Cuarentena/organización & administración , SARS-CoV-2 , Nave Espacial , Texas , Estados Unidos , United States National Aeronautics and Space Administration
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA