Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Life Sci Space Res (Amst) ; 31: 29-33, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34689947

RESUMO

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.


Assuntos
Voo Espacial , Ausência de Peso , Astronautas , Humanos , Sistemas Automatizados de Assistência Junto ao Leito , Tecnologia , Ausência de Peso/efeitos adversos
3.
J Immunol Methods ; 492: 112998, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33600819

RESUMO

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.


Assuntos
Citocinas/análise , Imunofenotipagem/métodos , Proteínas de Membrana/análise , Linfócitos T Reguladores/imunologia , Brefeldina A/farmacologia , Células Cultivadas , Citocinas/metabolismo , Enterotoxinas/imunologia , Voluntários Saudáveis , Humanos , Ativação Linfocitária , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Monensin/farmacologia , Cultura Primária de Células , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo
4.
J Allergy Clin Immunol Pract ; 8(10): 3247-3250, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32971311

RESUMO

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.


Assuntos
Astronautas , Infecções por Coronavirus/prevenção & controle , Hospedeiro Imunocomprometido/imunologia , Pandemias/prevenção & controle , Pneumonia Viral/prevenção & controle , Quarentena/métodos , Voo Espacial , Estresse Fisiológico/imunologia , Betacoronavirus , Contagem de Linfócito CD4 , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , COVID-19 , Infecções por Coronavirus/imunologia , Suplementos Nutricionais , Terapia por Exercício , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Humanos , Imunoglobulina G/uso terapêutico , Interleucina-2/uso terapêutico , Política Organizacional , Pneumonia Viral/imunologia , Quarentena/organização & administração , SARS-CoV-2 , Astronave , Texas , Estados Unidos , United States National Aeronautics and Space Administration
5.
Neurosci Biobehav Rev ; 115: 68-76, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32464118

RESUMO

The International Space Station (ISS) has continued to evolve from an operational perspective and multiple studies have monitored both stress and the immune system of ISS astronauts. Alterations were ascribed to a potentially synergistic array of factors, including microgravity, radiation, psychological stress, and circadian misalignment. Comparing similar data across 12 years of ISS construction and operations, we report that immunity, stress, and the reactivation of latent herpesviruses have all improved in ISS astronauts. Major physiological improvements seem to have initiated approximately 2012, a period coinciding with improvements onboard ISS including cargo delivery and resupply frequency, personal communication, exercise equipment and protocols, food quality and variety, nutritional supplementation, and schedule management. We conclude that spaceflight associated immune dysregulation has been positively influenced by operational improvements and biomedical countermeasures onboard ISS. Although an operational challenge, agencies should therefore incorporate, within vehicle design limitations, these dietary, operational, and stress-relieving countermeasures into deep space mission planning. Specific countermeasures that have benefited astronauts could serve as a therapy augment for terrestrial acquired immunodeficiency patients.


Assuntos
Herpesviridae , Voo Espacial , Astronautas , Humanos , Sistema Imunitário , Estresse Psicológico
6.
Life Sci Space Res (Amst) ; 25: 119-128, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32414485

RESUMO

BACKGROUND: On long-duration spaceflight, most astronauts experience persistent immune dysregulation and the reactivation of latent herpesviruses, including varicella zoster virus (VZV). To understand the clinical risk of these perturbations to astronauts, we paralleled the immunology and virology work-up of astronauts to otherwise healthy terrestrial persons with acute herpes zoster. METHODS: Blood samples from 42 zoster patients - confirmed positive by PCR for VZV DNA in saliva (range from 100 to >285 million copies/mL) were analyzed for peripheral leukocyte distribution, T cell function, and plasma cytokine profiles via multi-parametric flow cytometry and multiplex bead-based immune-array assays. Patient findings were compared to normal value ranges specific for each assay that were defined in-house previously from healthy adult test subjects. RESULTS: Compared to the healthy adult ranges, the zoster patients possess (1) a higher proportion of constitutively activated T-cells, (2) a T-cell population skewed towards a more experienced maturation state, (3) depressed general T-cell function, and (4) a higher concentration of 20 of 22 measured plasma cytokines. DISCUSSION: The pattern of immune dysregulation in zoster patients is similar to that of astronauts during spaceflight who shed VZV DNA in their saliva. Because future deep space exploration missions will be of an unprecedented duration, prolonged immune depression and chronic viral reactivation threaten to manifest overt disease in exploration class astronauts.


Assuntos
Citocinas/sangue , Herpes Zoster/imunologia , Herpesvirus Humano 3/fisiologia , Linfócitos T/imunologia , Adulto , Idoso , Astronautas , DNA Viral/análise , Feminino , Herpes Zoster/virologia , Herpesvirus Humano 3/imunologia , Herpesvirus Humano 3/isolamento & purificação , Humanos , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Saliva/virologia
7.
Hum Vaccin Immunother ; 16(12): 3138-3145, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32401699

RESUMO

The immunologic mechanisms underlying the improved serologic responses to heterologous prime-boost avian influenza vaccination are unclear. An exploratory analysis of the immune responses following 1 dose of influenza A/H7N9 inactivated vaccine in subjects who received an influenza A/H7N7 inactivated vaccine (N = 17) 8 years earlier or who were influenza A/H7-naïve (10) was performed. Plasma IL-6 and IL-21 concentrations by ELISA, the frequency of A/H7N7-specific memory B cells and antibody secreting cells by ELISpot, the frequency of circulating T follicular helper cells and the frequency of T cells expressing IL-6 and IL-21 by flow cytometry were assessed at baseline (D1), and 8 days (D9) and 28 days (D29) after vaccination. We assessed the correlation between these measurements and the D29 serologic responses to the boost vaccine. Plasma IL-6 concentration on D9 significantly correlated with the H7N7 and H7N9 hemagglutination inhibition (HAI) antibody levels (P = .03 and 0.02 respectively); and the percentage of T cells expressing IL-21 on D9 significantly correlated with H7N9 HAI antibody seroconversion (P < .001). Significant associations with other immunologic markers were not detected. We detected an association between plasma IL-6 and intracellular IL-21 and serologic responses to heterologous prime-boost avian influenza vaccination. A clarification of the role of these and additional immunologic markers requires larger clinical trials.


Assuntos
Imunidade Celular , Vírus da Influenza A Subtipo H7N7 , Subtipo H7N9 do Vírus da Influenza A , Vacinas contra Influenza , Influenza Aviária , Influenza Humana , Animais , Anticorpos Antivirais , Aves , Citocinas , Humanos , Vírus da Influenza A Subtipo H7N7/imunologia , Subtipo H7N9 do Vírus da Influenza A/imunologia , Influenza Humana/prevenção & controle , Vacinação , Vacinas de Produtos Inativados
8.
J Clin Invest ; 130(6): 3113-3123, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32125282

RESUMO

Mycobacterium tuberculosis (M. tuberculosis) has coevolved with humans for millennia and developed multiple mechanisms to evade host immunity. Restoring host immunity in order to improve outcomes and potentially shorten existing therapy will require identification of the full complement by which host immunity is inhibited. Perturbation of host DNA methylation is a mechanism induced by chronic infections such as HIV, HPV, lymphocytic choriomeningitis virus (LCMV), and schistosomiasis to evade host immunity. Here, we evaluated the DNA methylation status of patients with tuberculosis (TB) and their asymptomatic household contacts and found that the patients with TB have DNA hypermethylation of the IL-2/STAT5, TNF/NF-κB, and IFN-γ signaling pathways. We performed methylation-sensitive restriction enzyme-quantitative PCR (MSRE-qPCR) and observed that multiple genes of the IL-12/IFN-γ signaling pathway (IL12B, IL12RB2, TYK2, IFNGR1, JAK1, and JAK2) were hypermethylated in patients with TB. The DNA hypermethylation of these pathways was associated with decreased immune responsiveness with decreased mitogen-induced upregulation of IFN-γ, TNF, IL-6, CXCL9, CXCL10, and IL-1ß production. The DNA hypermethylation of the IL-12/IFN-γ pathway was associated with decreased IFN-γ-induced gene expression and decreased IL-12-inducible upregulation of IFN-γ. This study demonstrates that immune cells from patients with TB are characterized by DNA hypermethylation of genes critical to mycobacterial immunity resulting in decreased mycobacteria-specific and nonspecific immune responsiveness.


Assuntos
Metilação de DNA/imunologia , Regulação da Expressão Gênica/imunologia , Leucócitos/imunologia , Mycobacterium tuberculosis/imunologia , Transdução de Sinais/imunologia , Tuberculose/imunologia , Humanos , Leucócitos/patologia , Tuberculose/patologia
10.
Front Immunol ; 9: 1437, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30018614

RESUMO

Recent studies have established that dysregulation of the human immune system and the reactivation of latent herpesviruses persists for the duration of a 6-month orbital spaceflight. It appears certain aspects of adaptive immunity are dysregulated during flight, yet some aspects of innate immunity are heightened. Interaction between adaptive and innate immunity also seems to be altered. Some crews experience persistent hypersensitivity reactions during flight. This phenomenon may, in synergy with extended duration and galactic radiation exposure, increase specific crew clinical risks during deep space exploration missions. The clinical challenge is based upon both the frequency of these phenomena in multiple crewmembers during low earth orbit missions and the inability to predict which specific individual crewmembers will experience these changes. Thus, a general countermeasure approach that offers the broadest possible coverage is needed. The vehicles, architecture, and mission profiles to enable such voyages are now under development. These include deployment and use of a cis-Lunar station (mid 2020s) with possible Moon surface operations, to be followed by multiple Mars flyby missions, and eventual human Mars surface exploration. Current ISS studies will continue to characterize physiological dysregulation associated with prolonged orbital spaceflight. However, sufficient information exists to begin consideration of both the need for, and nature of, specific immune countermeasures to ensure astronaut health. This article will review relevant in-place operational countermeasures onboard ISS and discuss a myriad of potential immune countermeasures for exploration missions. Discussion points include nutritional supplementation and functional foods, exercise and immunity, pharmacological options, the relationship between bone and immune countermeasures, and vaccination to mitigate herpes (and possibly other) virus risks. As the immune system has sentinel connectivity within every other physiological system, translational effects must be considered for all potential immune countermeasures. Finally, we shall discuss immune countermeasures in the context of their individualized implementation or precision medicine, based on crewmember specific immunological biases.

11.
J Immunol ; 201(1): 124-133, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29752313

RESUMO

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.


Assuntos
Ascaríase/imunologia , Ascaris lumbricoides/imunologia , Vacina BCG/imunologia , Metilação de DNA/genética , Schistosoma haematobium/imunologia , Esquistossomose/imunologia , Células Th1/imunologia , Animais , Proliferação de Células/fisiologia , Células Cultivadas , Humanos , Interferon gama/genética , Interferon gama/imunologia , Interleucina-4/biossíntese , Interleucina-4/genética , Receptores de Citocinas/genética , Fatores de Transcrição/genética , Tuberculose/imunologia
12.
JCI Insight ; 3(2)2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29367462

RESUMO

T cell receptor (TCR) affinity is a critical factor of Treg lineage commitment, but whether self-reactivity is a determining factor in peripheral Treg function remains unknown. Here, we report that a high degree of self-reactivity is crucial for tissue-specific Treg function in autoimmunity. Based on high expression of CD5, we identified a subset of self-reactive Tregs expressing elevated levels of T-bet, GITR, CTLA-4, and ICOS, which imparted significant protection from autoimmune diabetes. We observed that T-bet expression in Tregs, necessary for control of Th1 autoimmunity, could be induced in an IFNγ-independent fashion and, unlike in conventional T cells (Tconv), was strongly correlated with the strength of TCR signaling. The level of CD5 similarly identified human Tregs with an increased functional profile, suggesting that CD5hi Tregs may constitute an efficacious subpopulation appropriate for use in adoptive Treg therapies for treatment of inflammatory conditions. Overall, this work establishes an instrumental role of high TCR self-reactivity in driving Treg function.


Assuntos
Autoimunidade , Diabetes Mellitus Tipo 1/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas com Domínio T/metabolismo , Linfócitos T Reguladores/imunologia , Adulto , Animais , Antígenos CD5 , Células Cultivadas , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/terapia , Modelos Animais de Doenças , Feminino , Voluntários Saudáveis , Humanos , Imunoterapia Adotiva/métodos , Ilhotas Pancreáticas , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Cultura Primária de Células , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas com Domínio T/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/transplante , Adulto Jovem
13.
Immunol Res ; 65(5): 1031-1045, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28822075

RESUMO

Although cytotoxic T lymphocytes (CTLs) store perforin within cytoplasmic secretory granules for immediate use, perforin is synthesized anew within hours of TCR stimulation. Previously, we observed new perforin protein at an immunologic synapse independent of secretory lysosomes; herein, we aimed to determine how new perforin transits to the synapse if not via lytic granules. We analyzed antigen-specific human CTLs via imaging flow cytometry and high-resolution confocal microscopy, with attention to intracellular trafficking components and new perforin. The recycling endosome compartments identified by rab8, rab11a, rab4, and rab37 co-localized with new perforin, as well as the SNAREs vti1b and VAMP4. After ablating the function of the recycling endosome pathway, we observed a relative accumulation of new perforin in rab8 vesicles. The recycling endosome pathway may serve as an auxiliary intracellular route for the delivery of new perforin to an immunologic synapse in order to perpetuate a cytotoxic response.


Assuntos
Endossomos/metabolismo , Sinapses Imunológicas/metabolismo , Perforina/metabolismo , Vesículas Secretórias/metabolismo , Linfócitos T Citotóxicos/imunologia , Células Cultivadas , Citotoxicidade Imunológica , Humanos , Transporte Proteico , Proteínas Qb-SNARE/metabolismo , Proteínas R-SNARE/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
14.
PLoS One ; 12(8): e0181134, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28767726

RESUMO

Natural killer (NK) cells are critical in immune defense against infected, stressed or transformed cells. Their function is regulated by the heterogeneous expression of a wide array of surface receptors that shape its phenotypic diversity. Although NK cells develop in the bone marrow and secondary lymphoid tissues, substantive differentiation is apparent in the peripheral blood including known age-related variation. In order to gain greater insight into phenotypic and functional variation within peripheral blood NK cells across age groups, we used multi-parametric, polyfunctional flow cytometry to interrogate the NK cell variability in 20 healthy adults and 15 5-10, 11-15 and 16-20 year-old children. We found that the normative ranges in both adults and children displayed great inter-individual variation for most markers. While the expression of several receptors did not differ, among those that did, the majority of the differences existed between adults and the three pediatric groups, rather than among children of different ages. Interestingly, we also identified variation in the individual expression of some markers by sex and ethnicity. Combinatorial analysis of NK cell receptors revealed intermediate subsets between the CD56bright and CD56dim NK cells. Furthermore, on examining the NK cell diversity by age, adults were discovered to have the lowest developmental diversity. Thus, our findings identify previously unappreciated NK cell subsets potentially distinguishing children from adults and suggest functional correlates that may have relevance in age-specific host defense.


Assuntos
Imunofenotipagem , Células Matadoras Naturais/metabolismo , Adolescente , Adulto , Fatores Etários , Antígeno CD56/metabolismo , Criança , Análise por Conglomerados , Feminino , Citometria de Fluxo , Humanos , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Leucócitos Mononucleares/citologia , Pessoa de Meia-Idade , Receptores de Células Matadoras Naturais/metabolismo , Adulto Jovem
15.
Front Pediatr ; 5: 104, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28555177

RESUMO

BACKGROUND: Several studies suggest that defects of regulatory T-cells (Tregs) and impaired cellular immunity are secondary to an imbalance between auto-aggressive T-cells and Tregs in lupus patients. Discrepancies in Tregs and effector T-cells (Teff) in active lupus patients are shown to be restored in patients upon receiving immunosuppressive therapy. Therefore, our main aim was to observe frequencies of these CD4+ T-cell subsets and Tregs/Teff ratio in a new diagnosis of childhood-onset systemic lupus erythematous (cSLE) before and after initiation of therapy. In addition, we monitored T-cell exhaustion status by examining responses to super-antigen staphylococcal enterotoxin B (SEB) and PD-1 expression in this patient. METHODS: Phenotyping of CD4+ T-cell subsets was carried out under basal conditions and after SEB stimulation using flow cytometry in one inactive (I-cSLE) and one active cSLE (A-cSLE) patient, as well as a healthy control (HC). The A-cSLE patient was a new diagnosis. Variables were measured at three consecutive time points in the active patient, reflecting various stages of disease activity. Activation status of CD4+ T-cells in the A-cSLE patient was compared to that of the I-cSLE patient and HC. Disease activity was measured by calculating the systemic lupus erythematous disease activity index. RESULTS: We found that the A-cSLE patient was not Tregs deficient. The patient had increased frequency of Tregs, and the Tregs/Teff ratio increased when the disease activity became less severe. CD4+ T-cells in the I-cSLE patient and in the A-cSLE patient with milder disease activity had heightened responsiveness to SEB, whereas T-cells were relatively hypo-responsive to SEB in the A-cSLE patient when disease activity was higher. The active patient exhibited higher frequencies of PD-1+ expressing Tregs, Teff, and Tnaïve/mem cells under basal conditions compared to the HC and I-cSLE patient. CONCLUSION: In the A-cSLE patient, changes in Tregs/Teff ratio correlated better with clinical improvement compared to Tregs frequencies alone and might reflect the restoration of immune homeostasis with therapy. SEB hypo-responsiveness in the A-cSLE patient when disease activity was higher paralleled with findings of greater frequencies of PD-1+ expressing Tregs, Teff, and Tnaïve/mem cells, suggests a possible global exhaustion status of CD4+ T-cells in this patient.

16.
J Immunol Methods ; 447: 47-51, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28434981

RESUMO

Modern immunologic studies demand increasing complexity because of a need to improve our understanding of the relationship between a cell's phenotype and its function. Regulatory T cells (Tregs) have been defined by a narrow set of phenotypic markers, however their actual functional capacity has not been determined at the single-cell level. Although the lymphocyte activation gene 3 (LAG-3; CD223) is a key marker for the identification of exhausted T cells, it may be useful also in resolving Treg subpopulations by indicating distinct functional breadths. Here we define the experimental conditions necessary for the optimal detection by flow cytometry of LAG-3 expression on activated Tregs. We stimulated human PBMCs with either PMA/ionomycin or Staphylococcal Enterotoxin B (SEB) and analyzed CD4+CD25+FoxP3+ Tregs for LAG-3 expression in concert with other Treg phenotypic markers. We prescribe a 24-hour stimulation period for the optimal detection of LAG-3 on Tregs. Furthermore, we determine LAG-3 protein expression on Tregs is compromised when the cells are treated with brefeldin A (BFA) and monensin. Therefore, the simultaneous assessment of Treg phenotype and function is complicated by the use of protein transport inhibitors.


Assuntos
Antígenos CD/genética , Brefeldina A/farmacologia , Monensin/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Antígenos CD/imunologia , Biomarcadores , Células Cultivadas , Regulação para Baixo , Enterotoxinas/farmacologia , Citometria de Fluxo , Humanos , Imunofenotipagem , Ionomicina/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Ativação Linfocitária , Transporte Proteico , Proteína do Gene 3 de Ativação de Linfócitos
17.
Blood ; 128(26): 3061-3072, 2016 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-27799162

RESUMO

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.


Assuntos
Síndrome de Job/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Citocinas/farmacologia , Proteínas de Ligação a DNA/metabolismo , Diterpenos/farmacologia , Meia-Vida , Fatores de Transcrição de Choque Térmico , Herpesvirus Humano 4/fisiologia , Humanos , Interleucina-17/metabolismo , Síndrome de Job/patologia , Camundongos , Modelos Moleculares , Proteínas Mutantes/metabolismo , Mutação/genética , Fosfotirosina/metabolismo , Ligação Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Fator de Transcrição STAT3/química , Fator de Transcrição STAT3/genética , Baço/patologia , Fatores de Transcrição/metabolismo
18.
PLoS Pathog ; 12(8): e1005805, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27486665

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proliferação de Células , Infecções por HIV/imunologia , Imunidade Celular , Adulto , Linfócitos T CD8-Positivos/patologia , Doença Crônica , Feminino , Infecções por HIV/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Perforina/imunologia , Proteínas com Domínio T/imunologia
19.
J Infect Dis ; 214(3): 479-88, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27389351

RESUMO

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.


Assuntos
Antígenos de Helmintos/metabolismo , Linfócitos T CD4-Positivos/efeitos dos fármacos , Fatores Imunológicos/metabolismo , Macrófagos/imunologia , Macrófagos/microbiologia , Mycobacterium tuberculosis/imunologia , Schistosoma/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Citocinas/biossíntese , Citometria de Fluxo , Humanos , Tolerância Imunológica , Imunofenotipagem , Leucócitos Mononucleares/imunologia , Lisossomos/metabolismo , Macrófagos/fisiologia , Fagossomos/metabolismo
20.
Mediators Inflamm ; 2016: 1478340, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27006526

RESUMO

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.


Assuntos
Imunidade Adaptativa/fisiologia , Infecções por HIV/complicações , Infecções por HIV/imunologia , Imunidade Inata/fisiologia , Mycobacterium tuberculosis/imunologia , Imunidade Adaptativa/genética , Adolescente , Adulto , Antígenos de Bactérias/imunologia , Complexo CD3/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Antígeno CD56/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Feminino , Humanos , Imunidade Inata/genética , Interferon gama/metabolismo , Interleucina-10/metabolismo , Interleucina-4/metabolismo , Masculino , Mycobacterium tuberculosis/patogenicidade , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...