Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Toxicol ; 42(1): 37-49, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36472205

RESUMO

Vaccines containing mRNA with the capacity to self-amplify represent an alternative to the mRNA vaccines that came to prominence during the COVID-19 pandemic. To gain further insights on the safety profile of self-amplifying mRNA- (SAM-) vaccines, this preclinical toxicology study in rats evaluated the effect of (i) the type of delivery system (lipid nanoparticle [LNP] vs cationic nano-emulsion [CNE]); (ii) antigen-encoding sequence (rabies glycoprotein G vs SARS-CoV-2 Spike); and (iii) RNA amplification. Further analyses also evaluated gene expression in peripheral blood after vaccination, and the biodistribution of vaccine RNA. The SAM vaccines administered as two doses 2-weeks apart had acceptable safety profiles in rats, with respect to clinical signs, blood biochemistry, and macroscopic and microscopic pathology. A transient increase in ALT/AST ratio occurred only in female rats and in the absence of muscle and liver damage was dependent on RNA amplification and appeared related to the greater quantities of vaccine RNA in the muscle and livers of female rats vs male rats. The RNA and delivery-vehicle components, but not the nature of the antigen-coding sequence or the requirement for RNA amplification, affected aspects of the stimulation of innate-immune activity, which was consistent with the transient activation of type I and type II interferon signaling. The delivery vehicle, LNP, differed from CNE as vaccine RNA in CNE compositions appeared independently to stimulate innate-immune activity at 4 hours after vaccination. Our analysis supports further studies to assess whether these differences in innate-immune activity affect safety and efficacy of the SAM vaccine.


Assuntos
COVID-19 , Vacinas , Ratos , Masculino , Feminino , Humanos , Animais , Pandemias , Distribuição Tecidual , COVID-19/prevenção & controle , SARS-CoV-2/genética , RNA Mensageiro , Vacinas Sintéticas
2.
Immunology ; 145(1): 161-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25521218

RESUMO

Extracellular signal-regulated kinase 3 (ERK3 )is an atypical member of the mitogen-activated protein kinase (MAPK) family. We have previously shown that ERK3 is expressed during thymocyte differentiation and that its expression is induced in mature peripheral T cells following activation of ERK1/2 by T-cell receptor (TCR) signalling. Herein, we have investigated whether ERK3 expression is required for proper T-cell selection. Using a knock-in mouse model in which the coding sequence of ERK3 is replaced by the gene encoding for the ß-galactosidase reporter, we show that ERK3 is expressed by double-positive (DP) thymocytes undergoing positive selection. In ERK3-deficient mice with a polyclonal TCR repertoire, we observe a decrease in positive selection. This reduction in positive selection was also observed when ERK3-deficient mice were backcrossed to class I- and class II-restricted TCR transgenic mice. Furthermore, the response of DP thymocytes to in vitro TCR stimulation was strongly reduced in ERK3-deficient mice. Together, these results show that ERK3 expression following TCR signalling is critical for proper thymic positive selection.


Assuntos
Seleção Clonal Mediada por Antígeno , Sistema de Sinalização das MAP Quinases/imunologia , Proteína Quinase 6 Ativada por Mitógeno/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Timócitos/imunologia , Timo/imunologia , Animais , Regulação Enzimológica da Expressão Gênica/genética , Regulação Enzimológica da Expressão Gênica/imunologia , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Knockout , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Proteína Quinase 6 Ativada por Mitógeno/genética , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/citologia , Timócitos/citologia , Timo/citologia
3.
J Antimicrob Chemother ; 67(12): 2919-26, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22899803

RESUMO

OBJECTIVES: To evaluate the relative propensities of oritavancin and vancomycin to induce Clostridium difficile infection (CDI) in hamster and in vitro human gut models. METHODS: Hamsters received clindamycin (100 mg/kg orally or subcutaneously), oritavancin (50 mg/kg orally) or vancomycin (50 mg/kg orally). C. difficile spores were administered orally the next day. Control hamsters received vehicle only (polyethylene glycol 400) plus spores or clindamycin but no spores. Hamsters were monitored for clinical signs for 20 days. Caecal contents were analysed for C. difficile cells, spores and the presence of (cyto)toxin. Oritavancin and vancomycin were instilled over 7 days into separate in vitro gut models primed with pooled human faeces and inoculated with C. difficile ribotype 027 spores. Gut flora, C. difficile total viable and spore counts, toxin titres and antimicrobial concentrations were determined. RESULTS: All hamsters treated with oritavancin survived up to 20 days, with no evidence of C. difficile spores, vegetative cells or toxin in their caeca. No hamsters treated with clindamycin or vancomycin survived >6 days after spore administration. Death was associated with high C. difficile counts and toxin in caecal contents. In the gut model, oritavancin dosing elicited a rapid, marked decrease in total viable C. difficile and spore counts to below the limit of detection. Vancomycin did not elicit germination or toxin production in the gut model, but C. difficile remained present as spores throughout. CONCLUSIONS: Oritavancin exposure, unlike exposure to vancomycin or clindamycin, did not lead to CDI in hamsters. In both models, oritavancin reduced C. difficile total counts and spores to below detectable limits. The data indicate the potential of oritavancin for CDI treatment, since exposure did not induce C. difficile germination and toxin production, which are known to exacerbate the disease state.


Assuntos
Antibacterianos/metabolismo , Toxinas Bacterianas/biossíntese , Clostridioides difficile/efeitos dos fármacos , Clostridioides difficile/crescimento & desenvolvimento , Glicopeptídeos/metabolismo , Esporos Bacterianos/efeitos dos fármacos , Esporos Bacterianos/crescimento & desenvolvimento , Idoso , Idoso de 80 Anos ou mais , Animais , Carga Bacteriana , Ceco/química , Ceco/microbiologia , Infecções por Clostridium/microbiologia , Infecções por Clostridium/patologia , Cricetinae , Modelos Animais de Doenças , Fezes/química , Fezes/microbiologia , Humanos , Lipoglicopeptídeos , Vancomicina/metabolismo
4.
J Virol ; 83(19): 9743-58, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19605470

RESUMO

We previously reported that CD4C/human immunodeficiency virus (HIV)(Nef) transgenic (Tg) mice, expressing Nef in CD4(+) T cells and cells of the macrophage/dendritic cell (DC) lineage, develop a severe AIDS-like disease, characterized by depletion of CD4(+) T cells, as well as lung, heart, and kidney diseases. In order to determine the contribution of distinct populations of hematopoietic cells to the development of this AIDS-like disease, five additional Tg strains expressing Nef through restricted cell-specific regulatory elements were generated. These Tg strains express Nef in CD4(+) T cells, DCs, and macrophages (CD4E/HIV(Nef)); in CD4(+) T cells and DCs (mCD4/HIV(Nef) and CD4F/HIV(Nef)); in macrophages and DCs (CD68/HIV(Nef)); or mainly in DCs (CD11c/HIV(Nef)). None of these Tg strains developed significant lung and kidney diseases, suggesting the existence of as-yet-unidentified Nef-expressing cell subset(s) that are responsible for inducing organ disease in CD4C/HIV(Nef) Tg mice. Mice from all five strains developed persistent oral carriage of Candida albicans, suggesting an impaired immune function. Only strains expressing Nef in CD4(+) T cells showed CD4(+) T-cell depletion, activation, and apoptosis. These results demonstrate that expression of Nef in CD4(+) T cells is the primary determinant of their depletion. Therefore, the pattern of Nef expression in specific cell population(s) largely determines the nature of the resulting pathological changes.


Assuntos
Síndrome da Imunodeficiência Adquirida/virologia , Regulação Viral da Expressão Gênica , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Animais , Antígenos CD/biossíntese , Antígenos de Diferenciação Mielomonocítica/biossíntese , Antígeno CD11c/biossíntese , Antígenos CD4/biossíntese , Linfócitos T CD4-Positivos/virologia , Separação Celular , Células Dendríticas/virologia , Modelos Animais de Doenças , Humanos , Macrófagos/virologia , Camundongos , Camundongos Transgênicos
5.
Appl Health Econ Health Policy ; 17(6): 827-839, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31392669

RESUMO

BACKGROUND: Current strategies for risk stratification of patients with acute myeloid leukemia assign approximately 40% of patients to the intermediate-risk group, where uncertainty about optimal therapy still persists. OBJECTIVE: The objective of this study was to assess the cost effectiveness of a HMGA2 prognostic test based on HMGA2+/HMGA2- expression, which improves genetic risk stratification in acute myeloid leukemia, and compare this test with the current standard of care in Canada. METHODS: A cost-effectiveness model was developed from the Canadian National Healthcare Service and societal perspective using data from the Quebec Leukemia Cell Bank, published literature, and physician surveys. The model includes a lifetime horizon assessing the HMGA2 test vs. standard of care. RESULTS: The HMGA2 test outperformed the standard of care at all time horizons culminating with estimated improvements of 1.92 and 3.12 months in leukemia-free survival and overall survival, respectively. Costs associated with the HMGA2 test were consistently lower, except diagnostic costs, routine medical costs, and costs related to infections and false positives. From a societal perspective, total lifetime costs were $161,358 CAD and $151,908 CAD with the standard of care and the HMGA2 test, respectively. The incremental quality-adjusted life-year gain was 0.138, which led to dominance over the standard of care. Deterministic sensitivity analyses confirmed the results of the base-case scenario. Probabilistic sensitivity analyses revealed that for a willingness-to-pay threshold of $100,000 CAD, the probability of cost effectiveness was 87.19%. CONCLUSIONS: The HMGA2 test is estimated to improve leukemia-free survival and overall survival outcomes, and yield costs savings from a healthcare system and societal perspective.


Assuntos
Análise Custo-Benefício , Técnicas e Procedimentos Diagnósticos/economia , Proteína HMGA2/análise , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/fisiopatologia , Biomarcadores/análise , Canadá , Humanos , Leucemia Mieloide Aguda/terapia , Prognóstico , Anos de Vida Ajustados por Qualidade de Vida
6.
Blood Cancer J ; 9(3): 28, 2019 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-30820024

RESUMO

Since the publication of the original article the authors noticed the the affiliation details for Paresh Vyas are incorrect. The correct affiliation details for this author are given below.

7.
J Leukoc Biol ; 82(3): 645-56, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17554014

RESUMO

IL-21 is a Type I cytokine, which uses the common gamma chain (gamma(c)) in its receptor. As members of the gamma(c) cytokine/cytokine receptors family play crucial role in the differentiation, activation, and survival of lymphocytes, we have investigated if IL-21 could promote T cell survival and thus, contribute to T cell homeostasis and expansion. Unlike most gamma(c) cytokine receptors, we report that IL-21R is constitutively expressed by all mature T lymphocytes and that stromal cells of lymphoid organs are a constitutive source of IL-21. These observations are reminiscent of what is observed for IL-7/IL-7R, which control T cell survival and homeostasis and suggest a role for IL-21 in T cell homeostasis. Indeed, our results show that IL-21 is a survival factor for resting and activated T cells. Moreover, the ability of IL-21 to costimulate T cell proliferation is mediated by enhancing T cell viability. Further investigation of how IL-21R signaling induces T cell survival shows for the first time that IL-21 binding to its receptor activates the PI-3K signaling pathway and induces Bcl-2 expression. Moreover, the activation of the PI-3K signaling pathway is essential for IL-21-mediated T cell survival. Our data provide a new role for IL-21 in the immune system, which might be used to improve T cell homeostasis in immunocompromised patients.


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Interleucinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Linfócitos T/citologia , Animais , Western Blotting , Tamanho Celular , Ativação Enzimática , Citometria de Fluxo , Subunidade alfa de Receptor de Interleucina-21/genética , Subunidade alfa de Receptor de Interleucina-21/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Linfócitos T/metabolismo
8.
Leukemia ; 32(6): 1349-1357, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29550835

RESUMO

Acute promyelocytic leukemia (APL) is a medical emergency because of associated lethal early bleeding, a condition preventable by prompt diagnosis and therapeutic intervention. The mechanisms underlying the hemostatic anomalies of APL are not completely elucidated. RNA-sequencing-based characterization of APL (n = 30) was performed and compared to that of other acute myeloid leukemia (n = 400) samples and normal promyelocytes. Perturbations in the transcriptome of coagulation and fibrinolysis-related genes in APL extend beyond known culprits and now include Thrombin, Factor X and Urokinase Receptor. Most intriguingly, the Podoplanin (PDPN) gene, involved in platelet aggregation, is aberrantly expressed in APL promyelocytes and is the most distinctive transcript for this disease. Using an antibody panel optimized for AML diagnosis by flow cytometry, we also found that PDPN was the most specific surface marker for APL, and that all-trans retinoic acid therapy rapidly decreases its expression. Functional studies showed that engineered overexpression of this gene in human leukemic cells causes aberrant platelet binding, activation and aggregation. PDPN-expressing primary APL cells, but not PDPN-negative primary leukemias, specifically induce platelet binding, activation and aggregation. Finally, PDPN expression on leukemia cells in a xenograft model was associated with thrombocytopenia and prolonged bleeding time in vivo. Together our results suggest that PDPN may contribute to the hemostatic perturbations found in APL.


Assuntos
Hemorragia/etiologia , Leucemia Promielocítica Aguda/complicações , Glicoproteínas de Membrana/fisiologia , Transcriptoma , Adulto , Idoso , Animais , Feminino , Citometria de Fluxo , Humanos , Leucemia Promielocítica Aguda/genética , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Pessoa de Meia-Idade , Agregação Plaquetária , Trombocitopenia/etiologia , Tretinoína/farmacologia
9.
Blood Cancer J ; 8(8): 68, 2018 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-30061630

RESUMO

In acute myeloid leukemia (AML), risk stratification based on cytogenetics and mutation profiling is essential but remains insufficient to select the optimal therapy. Accurate biomarkers are needed to improve prognostic assessment. We analyzed RNA sequencing and survival data of 430 AML patients and identified HMGA2 as a novel prognostic marker. We validated a quantitative PCR test to study the association of HMGA2 expression with clinical outcomes in 358 AML samples. In this training cohort, HMGA2 was highly expressed in 22.3% of AML, mostly in patients with intermediate or adverse cytogenetics. High expression levels of HMGA2 (H + ) were associated with a lower frequency of complete remission (58.8% vs 83.4%, P < 0.001), worse 3-year overall survival (OS, 13.2% vs 43.5%, P < 0.001) and relapse-free survival (RFS, 10.8% vs 44.2%, P < 0.001). A positive HMGA2 test also identified a subgroup of patients unresponsive to standard treatments. Multivariable analyses showed that H + was independently associated with significantly worse OS and RFS, including in the intermediate cytogenetic risk category. These associations were confirmed in a validation cohort of 260 patient samples from the UK NCRI AML17 trial. The HMGA2 test could be implemented in clinical trials developing novel therapeutic strategies for high-risk AML.

10.
Mol Cell Biol ; 34(18): 3374-87, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25002529

RESUMO

Extracellular signal-regulated kinase 3 (ERK3) is an atypical member of the mitogen-activated protein kinase (MAPK) family whose function is largely unknown. Given the central role of MAPKs in T cell development, we hypothesized that ERK3 may regulate thymocyte development. Here we have shown that ERK3 deficiency leads to a 50% reduction in CD4(+) CD8(+) (DP) thymocyte number. Analysis of hematopoietic chimeras revealed that the reduction in DP thymocytes is intrinsic to hematopoietic cells. We found that early thymic progenitors seed the Erk3(-/-) thymus and can properly differentiate and proliferate to generate DP thymocytes. However, ERK3 deficiency results in a decrease in the DP thymocyte half-life, associated with a higher level of apoptosis. As a consequence, ERK3-deficient DP thymocytes are impaired in their ability to make successful secondary T cell receptor alpha (TCRα) gene rearrangement. Introduction of an already rearranged TCR transgene restores thymic cell number. We further show that knock-in of a catalytically inactive allele of Erk3 fails to rescue the loss of DP thymocytes. Our results uncover a unique role for ERK3, dependent on its kinase activity, during T cell development and show that this atypical MAPK is essential to sustain DP survival during RAG-mediated rearrangements.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteína Quinase 6 Ativada por Mitógeno/genética , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Timócitos/citologia , Timo/citologia , Animais , Animais Recém-Nascidos , Linfócitos T CD4-Positivos/enzimologia , Linfócitos T CD8-Positivos/enzimologia , Domínio Catalítico , Diferenciação Celular/genética , Proliferação de Células , Sobrevivência Celular , Embrião de Mamíferos , Técnicas de Introdução de Genes , Rearranjo Gênico da Cadeia alfa dos Receptores de Antígenos dos Linfócitos T , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Timócitos/imunologia
11.
PLoS One ; 9(1): e86681, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24475167

RESUMO

The classical mitogen-activated protein kinases (MAPKs) ERK1 and ERK2 are activated upon stimulation of cells with a broad range of extracellular signals (including antigens) allowing cellular responses to occur. ERK3 is an atypical member of the MAPK family with highest homology to ERK1/2. Therefore, we evaluated the role of ERK3 in mature T cell response. Mouse resting T cells do not transcribe ERK3 but its expression is induced in both CD4⁺ and CD8⁺ T cells following T cell receptor (TCR)-induced T cell activation. This induction of ERK3 expression in T lymphocytes requires activation of the classical MAPK ERK1 and ERK2. Moreover, ERK3 protein is phosphorylated and associates with MK5 in activated primary T cells. We show that ERK3-deficient T cells have a decreased proliferation rate and are impaired in cytokine secretion following in vitro stimulation with low dose of anti-CD3 antibodies. Our findings identify the atypical MAPK ERK3 as a new and important regulator of TCR-induced T cell activation.


Assuntos
Regulação da Expressão Gênica/imunologia , Imunidade Celular/imunologia , Ativação Linfocitária/imunologia , Proteína Quinase 6 Ativada por Mitógeno/metabolismo , Linfócitos T/imunologia , Animais , Proliferação de Células , Citocinas/metabolismo , Primers do DNA/genética , Citometria de Fluxo , Immunoblotting , Imunoprecipitação , Camundongos , Proteína Quinase 6 Ativada por Mitógeno/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/metabolismo , beta-Galactosidase
12.
Eur J Immunol ; 37(11): 3069-77, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17918202

RESUMO

The ability of IL-21 to promote in vitro T cell survival led us to investigate its biological activity in vivo. We report that overexpression of IL-21 in transgenic mice drives CD8(+) memory T cell accumulation with a concomitant reduction in naive T cell numbers. These memory T cells are functional, given their ability to rapidly produce IFN-gamma and proliferate following stimulation. Since the homeostasis of naive and memory T cells is controlled by cytokines, we evaluated whether IL-21 influences cytokine receptor expression. We show that IL-21 inhibits IL-7R expression on naive T cells in vitro, suggesting impaired IL-7-mediated naive T cell survival in IL-21-transgenic mice. In contrast, IL-7R expression on CD4(+) memory T cells is not affected, allowing their IL-7-dependent survival in IL-21-transgenic mice. Although IL-21 decreases IL-7R expression on CD8(+) memory T cells, this has no impact on their survival since their maintenance in the T cell pool is IL-7-independent. Rather, we demonstrate that CD8(+) memory T cells are receptive to IL-21 survival signals allowing for their accumulation in IL-21-transgenic mice. This study identifies new roles for IL-21 in T cell homeostasis and in the regulation of T cell responses to cytokines.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Memória Imunológica , Interleucinas/metabolismo , Subpopulações de Linfócitos T/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Citometria de Fluxo , Interleucina-7/imunologia , Interleucina-7/metabolismo , Interleucinas/imunologia , Camundongos , Camundongos Transgênicos , Receptores de Interleucina-7/imunologia , Receptores de Interleucina-7/metabolismo , Subpopulações de Linfócitos T/imunologia
13.
Infect Immun ; 74(4): 2382-91, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16552068

RESUMO

Candida albicans causes oropharyngeal candidiasis (OPC) but rarely disseminates to deep organs in human immunodeficiency virus (HIV) infection. Here, we used a model of OPC in CD4C/HIV(Mut) transgenic (Tg) mice to investigate the role of polymorphonuclear leukocytes (PMNs) and CD8+ T cells in limiting candidiasis to the mucosa. Numbers of circulating PMNs and their oxidative burst were both augmented in CD4C/HIV(MutA) Tg mice expressing rev, env, and nef of HIV type 1 (HIV-1), while phagocytosis and killing of C. albicans were largely unimpaired compared to those in non-Tg mice. Depletion of PMNs in these Tg mice did not alter oral or gastrointestinal burdens of C. albicans or cause systemic dissemination. However, oral burdens of C. albicans were increased in CD4C/HIV(MutG) Tg mice expressing only the nef gene of HIV-1 and bred on a CD8 gene-deficient background (CD8-/-), compared to control or heterozygous CD8+/- CD4C/HIV(MutG) Tg mice. Thus, CD8+ T cells contribute to the host defense against oral candidiasis in vivo, specifically in the context of nef expression in a subset of immune cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/microbiologia , Candida albicans/imunologia , Candidíase Bucal/imunologia , Infecções por HIV/imunologia , HIV-1/genética , Neutrófilos/imunologia , Neutrófilos/microbiologia , Animais , Antígenos CD4/genética , Antígenos CD8/genética , Linfócitos T CD8-Positivos/virologia , Candidíase Bucal/genética , Candidíase Bucal/virologia , Células Cultivadas , Doença Crônica , Modelos Animais de Doenças , Feminino , Infecções por HIV/genética , Infecções por HIV/microbiologia , Humanos , Leucopenia/genética , Leucopenia/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Camundongos Transgênicos , Neutrófilos/virologia , Fagocitose/genética , Explosão Respiratória/genética , Fatores de Tempo
14.
J Immunol ; 177(1): 479-91, 2006 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-16785545

RESUMO

The impairments of protective mucosal immunity which cause susceptibility to oropharyngeal candidiasis (OPC) in HIV infection remain undefined. This study used a model of OPC in CD4C/HIV MutA transgenic (Tg) mice expressing Rev, Env, and Nef of HIV-1 to investigate the role of transgene expressing dendritic cells (DCs) and CD4+ T cells in maintenance of chronic oral carriage of Candida albicans. DCs were depleted in the Tg mice and had an immature phenotype, with low expression of MHC class II and IL-12. CD4+ T cells were quantitatively reduced in the oral mucosa, cervical lymph nodes (CLNs) and peripheral blood of the Tg mice, and displayed a polarization toward a nonprotective Th2 response. Proliferation of CLN CD4+ T cells from infected Tg mice in response to C. albicans Ag in vitro was abrogated and the cells failed to acquire an effector phenotype. Coculture of C. albicans-pulsed DCs with CD4+ T cells in vitro showed that Tg expression in either or both of these cell populations sharply reduced the proliferation of CD4+ T cells and their production of IL-2. Finally, transfer of naive non-Tg CD4+ T cells into these Tg mice restored proliferation to C. albicans Ag and sharply reduced oral burdens of C. albicans. Overall, these results indicate that defective CD4+ T cells primarily determine the susceptibility to chronic carriage of C. albicans in these Tg mice.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Candidíase/genética , Candidíase/imunologia , Predisposição Genética para Doença , HIV-1/genética , HIV-1/imunologia , Imunofenotipagem , Mucosa Bucal/imunologia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/microbiologia , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/microbiologia , Linfócitos T CD8-Positivos/virologia , Candida albicans/imunologia , Candidíase/patologia , Candidíase/virologia , Proliferação de Células , Técnicas de Cocultura , Citocinas/biossíntese , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Células Dendríticas/transplante , Células Dendríticas/virologia , Feminino , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/microbiologia , Infecções por HIV/patologia , Imunidade nas Mucosas/genética , Memória Imunológica/genética , Linfonodos/imunologia , Linfonodos/microbiologia , Linfonodos/patologia , Linfonodos/virologia , Ativação Linfocitária/genética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Transgênicos , Mucosa Bucal/microbiologia , Mucosa Bucal/virologia , Pescoço , Fagocitose/genética , Fagocitose/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA