Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
N Engl J Med ; 390(17): 1549-1559, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38669354

RESUMO

BACKGROUND: Subcutaneous administration of the monoclonal antibody L9LS protected adults against controlled Plasmodium falciparum infection in a phase 1 trial. Whether a monoclonal antibody administered subcutaneously can protect children from P. falciparum infection in a region where this organism is endemic is unclear. METHODS: We conducted a phase 2 trial in Mali to assess the safety and efficacy of subcutaneous administration of L9LS in children 6 to 10 years of age over a 6-month malaria season. In part A of the trial, safety was assessed at three dose levels in adults, followed by assessment at two dose levels in children. In part B of the trial, children were randomly assigned, in a 1:1:1 ratio, to receive 150 mg of L9LS, 300 mg of L9LS, or placebo. The primary efficacy end point, assessed in a time-to-event analysis, was the first P. falciparum infection, as detected on blood smear performed at least every 2 weeks for 24 weeks. A secondary efficacy end point was the first episode of clinical malaria, as assessed in a time-to-event analysis. RESULTS: No safety concerns were identified in the dose-escalation part of the trial (part A). In part B, 225 children underwent randomization, with 75 children assigned to each group. No safety concerns were identified in part B. P. falciparum infection occurred in 36 participants (48%) in the 150-mg group, in 30 (40%) in the 300-mg group, and in 61 (81%) in the placebo group. The efficacy of L9LS against P. falciparum infection, as compared with placebo, was 66% (adjusted confidence interval [95% CI], 45 to 79) with the 150-mg dose and 70% (adjusted 95% CI, 50 to 82) with the 300-mg dose (P<0.001 for both comparisons). Efficacy against clinical malaria was 67% (adjusted 95% CI, 39 to 82) with the 150-mg dose and 77% (adjusted 95% CI, 55 to 89) with the 300-mg dose (P<0.001 for both comparisons). CONCLUSIONS: Subcutaneous administration of L9LS to children was protective against P. falciparum infection and clinical malaria over a period of 6 months. (Funded by the National Institute of Allergy and Infectious Diseases; ClinicalTrials.gov number, NCT05304611.).


Assuntos
Anticorpos Monoclonais Humanizados , Malária Falciparum , Adulto , Criança , Feminino , Humanos , Masculino , Relação Dose-Resposta a Droga , Método Duplo-Cego , Doenças Endêmicas/prevenção & controle , Injeções Subcutâneas , Estimativa de Kaplan-Meier , Malária Falciparum/tratamento farmacológico , Malária Falciparum/epidemiologia , Malária Falciparum/prevenção & controle , Mali/epidemiologia , Plasmodium falciparum , Resultado do Tratamento , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Terapia Diretamente Observada , Combinação Arteméter e Lumefantrina/administração & dosagem , Combinação Arteméter e Lumefantrina/uso terapêutico , Adulto Jovem , Pessoa de Meia-Idade
2.
Sci Transl Med ; 15(692): eade4790, 2023 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-37075129

RESUMO

Influenza vaccines could be improved by platforms inducing cross-reactive immunity. Immunodominance of the influenza hemagglutinin (HA) head in currently licensed vaccines impedes induction of cross-reactive neutralizing stem-directed antibodies. A vaccine without the variable HA head domain has the potential to focus the immune response on the conserved HA stem. This first-in-human dose-escalation open-label phase 1 clinical trial (NCT03814720) tested an HA stabilized stem ferritin nanoparticle vaccine (H1ssF) based on the H1 HA stem of A/New Caledonia/20/1999. Fifty-two healthy adults aged 18 to 70 years old enrolled to receive either 20 µg of H1ssF once (n = 5) or 60 µg of H1ssF twice (n = 47) with a prime-boost interval of 16 weeks. Thirty-five (74%) 60-µg dose participants received the boost, whereas 11 (23%) boost vaccinations were missed because of public health restrictions in the early stages of the COVID-19 pandemic. The primary objective of this trial was to evaluate the safety and tolerability of H1ssF, and the secondary objective was to evaluate antibody responses after vaccination. H1ssF was safe and well tolerated, with mild solicited local and systemic reactogenicity. The most common symptoms included pain or tenderness at the injection site (n = 10, 19%), headache (n = 10, 19%), and malaise (n = 6, 12%). We found that H1ssF elicited cross-reactive neutralizing antibodies against the conserved HA stem of group 1 influenza viruses, despite previous H1 subtype head-specific immunity. These responses were durable, with neutralizing antibodies observed more than 1 year after vaccination. Our results support this platform as a step forward in the development of a universal influenza vaccine.


Assuntos
COVID-19 , Vacinas contra Influenza , Influenza Humana , Adolescente , Adulto , Idoso , Humanos , Pessoa de Meia-Idade , Adulto Jovem , Anticorpos Neutralizantes , Anticorpos Antivirais , Anticorpos Amplamente Neutralizantes , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Hemaglutininas , Pandemias
3.
Nat Med ; 28(5): 1022-1030, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35411076

RESUMO

Adeno-associated viral vector-mediated transfer of DNA coding for broadly neutralizing anti-HIV antibodies (bnAbs) offers an alternative to attempting to induce protection by vaccination or by repeated infusions of bnAbs. In this study, we administered a recombinant bicistronic adeno-associated virus (AAV8) vector coding for both the light and heavy chains of the potent broadly neutralizing HIV-1 antibody VRC07 (AAV8-VRC07) to eight adults living with HIV. All participants remained on effective anti-retroviral therapy (viral load (VL) <50 copies per milliliter) throughout this phase 1, dose-escalation clinical trial ( NCT03374202 ). AAV8-VRC07 was given at doses of 5 × 1010, 5 × 1011 and 2.5 × 1012 vector genomes per kilogram by intramuscular (IM) injection. Primary endpoints of this study were to assess the safety and tolerability of AAV8-VRC07; to determine the pharmacokinetics and immunogenicity of in vivo VRC07 production; and to describe the immune response directed against AAV8-VRC07 vector and its products. Secondary endpoints were to assess the clinical effects of AAV8-VRC07 on CD4 T cell count and VL and to assess the persistence of VRC07 produced in participants. In this cohort, IM injection of AAV8-VRC07 was safe and well tolerated. No clinically significant change in CD4 T cell count or VL occurred during the 1-3 years of follow-up reported here. In participants who received AAV8-VRC07, concentrations of VRC07 were increased 6 weeks (P = 0.008) and 52 weeks (P = 0.016) after IM injection of the product. All eight individuals produced measurable amounts of serum VRC07, with maximal VRC07 concentrations >1 µg ml-1 in three individuals. In four individuals, VRC07 serum concentrations remained stable near maximal concentration for up to 3 years of follow-up. In exploratory analyses, neutralizing activity of in vivo produced VRC07 was similar to that of in vitro produced VRC07. Three of eight participants showed a non-idiotypic anti-drug antibody (ADA) response directed against the Fab portion of VRC07. This ADA response appeared to decrease the production of serum VRC07 in two of these three participants. These data represent a proof of concept that adeno-associated viral vectors can durably produce biologically active, difficult-to-induce bnAbs in vivo, which could add valuable new tools to the fight against infectious diseases.


Assuntos
Infecções por HIV , HIV-1 , Adulto , Anticorpos Neutralizantes , Anticorpos Amplamente Neutralizantes , Dependovirus/genética , Anticorpos Anti-HIV , Infecções por HIV/tratamento farmacológico , Humanos
4.
Cell Rep ; 22(4): 860-868, 2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29420173

RESUMO

Unequal transmission of nutritive signaling during cell division establishes fate disparity between sibling lymphocytes, but how asymmetric signaling becomes organized is not understood. We show that receptor-associated class I phosphatidylinositol 3-kinase (PI3K) signaling activity, indexed by phosphatidylinositol (3,4,5)-trisphosphate (PIP3) staining, is spatially restricted to the microtubule-organizing center and subsequently to one pole of the mitotic spindle in activated T and B lymphocytes. Asymmetric PI3K activity co-localizes with polarization of antigen receptor components implicated in class I PI3K signaling and with facultative glucose transporters whose trafficking is PI3K dependent and whose abundance marks cells destined for differentiation. Perturbation of class I PI3K activity disrupts asymmetry of upstream antigen receptors and downstream glucose transporter traffic. The roles of PI3K signaling in nutrient utilization, proliferation, and gene expression may have converged with the conserved role of PI3K signaling in cellular symmetry breaking to form a logic for regenerative lymphocyte divisions.


Assuntos
Linfócitos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Diferenciação Celular , Humanos , Transdução de Sinais
6.
Immunohorizons ; 1(7): 156-161, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28944344

RESUMO

Anabolic metabolism in lymphocytes promotes plasmablast and cytotoxic T cell differentiation at the expense of self-renewal. Heightened expression and function of the transcription factor IFN regulatory factor 4 (IRF4) accompany enhanced anabolic induction and full commitment to functional differentiation in B cells and CD8+ T cells. In this study, we used a genetic approach to determine whether IRF4 plays an analogous role in Th1 cell induction. Our findings indicate that IRF4 promotes determined Th1 cell differentiation in tandem with anabolic metabolism of CD4+ T cells. IRF4-deficient CD4+ T cells stimulated in vitro exhibit impaired induction of Th1 gene expression and defective silencing of T cell factor 1 expression. IRF4-deficient CD4+ T cells also undergo a shift toward catabolic metabolism, with reduced mammalian target of rapamycin activation, cell size, and nutrient uptake, as well as increased mitochondrial clearance. These findings suggest that the ability to remodel metabolic states can be an essential gateway for altering cell fate.

7.
J Exp Med ; 214(1): 39-47, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27923906

RESUMO

Upon infection, an activated CD4+ T cell produces terminally differentiated effector cells and renews itself for continued defense. In this study, we show that differentiation and self-renewal arise as opposing outcomes of sibling CD4+ T cells. After influenza challenge, antigen-specific cells underwent several divisions in draining lymph nodes (LN; DLNs) while maintaining expression of TCF1. After four or five divisions, some cells silenced, whereas some cells maintained TCF1 expression. TCF1-silenced cells were T helper 1-like effectors and concentrated in the lungs. Cells from earliest divisions were memory-like and concentrated in nondraining LN. TCF1-expressing cells from later divisions in the DLN could self-renew, clonally yielding a TCF1-silenced daughter cell as well as a sibling cell maintaining TCF1 expression. Some TCF1-expressing cells in DLNs acquired an alternative, follicular helper-like fate. Modeled differentiation experiments in vitro suggested that unequal PI3K/mechanistic target of rapamycin signaling drives intraclonal cell fate heterogeneity. Asymmetric division enables self-renewal to be coupled to production of differentiated CD4+ effector T cells during clonal selection.


Assuntos
Divisão Celular Assimétrica/fisiologia , Linfócitos T CD4-Positivos/imunologia , Animais , Divisão Celular , Células Cultivadas , Fator 1-alfa Nuclear de Hepatócito/análise , Fator 1-alfa Nuclear de Hepatócito/genética , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/fisiologia , Serina-Treonina Quinases TOR/fisiologia
8.
Cell Rep ; 17(12): 3142-3152, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-28009285

RESUMO

Regeneration requires related cells to diverge in fate. We show that activated lymphocytes yield sibling cells with unequal elimination of aged mitochondria. Disparate mitochondrial clearance impacts cell fate and reflects larger constellations of opposing metabolic states. Differentiation driven by an anabolic constellation of PI3K/mTOR activation, aerobic glycolysis, inhibited autophagy, mitochondrial stasis, and ROS production is balanced with self-renewal maintained by a catabolic constellation of AMPK activation, mitochondrial elimination, oxidative metabolism, and maintenance of FoxO1 activity. Perturbations up and down the metabolic pathways shift the balance of nutritive constellations and cell fate owing to self-reinforcement and reciprocal inhibition between anabolism and catabolism. Cell fate and metabolic state are linked by transcriptional regulators, such as IRF4 and FoxO1, with dual roles in lineage and metabolic choice. Instructing some cells to utilize nutrients for anabolism and differentiation while other cells catabolically self-digest and self-renew may enable growth and repair in metazoa.


Assuntos
Proteína Forkhead Box O1/genética , Fatores Reguladores de Interferon/genética , Ativação Linfocitária/genética , Linfócitos/metabolismo , Mitocôndrias/metabolismo , Animais , Autofagia/genética , Diferenciação Celular/genética , Proteína Forkhead Box O1/metabolismo , Glicólise , Hematopoese/genética , Fatores Reguladores de Interferon/metabolismo , Metabolismo/genética , Camundongos , Mitocôndrias/genética , Fosfatidilinositol 3-Quinases/genética , Espécies Reativas de Oxigênio/metabolismo , Regeneração/genética , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética
9.
Cell Rep ; 17(7): 1773-1782, 2016 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-27829149

RESUMO

Selected CD8+ T cells must divide, produce differentiated effector cells, and self-renew, often repeatedly. We now show that silencing expression of the transcription factor TCF1 marks loss of self-renewal by determined effector cells and that this requires cell division. In acute infections, the first three CD8+ T cell divisions produce daughter cells with unequal proliferative signaling but uniform maintenance of TCF1 expression. The more quiescent initial daughter cells resemble canonical central memory cells. The more proliferative, effector-prone cells from initial divisions can subsequently undergo division-dependent production of a TCF1-negative effector daughter cell along with a self-renewing TCF1-positive daughter cell, the latter also contributing to the memory cell pool upon resolution of infection. Self-renewal in the face of effector cell determination may promote clonal amplification and memory cell formation in acute infections, sustain effector regeneration during persistent subclinical infections, and be rate limiting, but remediable, in chronic active infections and cancer.


Assuntos
Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Autorrenovação Celular , Animais , Ciclo Celular , Diferenciação Celular , Divisão Celular , Proliferação de Células , Células Clonais , Inativação Gênica , Camundongos Endogâmicos C57BL , Fator 1 de Transcrição de Linfócitos T/metabolismo
10.
Cell Rep ; 13(10): 2203-18, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26628372

RESUMO

Metazoan sibling cells often diverge in activity and identity, suggesting links between growth signals and cell fate. We show that unequal transduction of nutrient-sensitive PI3K/AKT/mTOR signaling during cell division bifurcates transcriptional networks and fates of kindred cells. A sibling B lymphocyte with stronger signaling, indexed by FoxO1 inactivation and IRF4 induction, undergoes PI3K-driven Pax5 repression and plasma cell determination, while its sibling with weaker PI3K activity renews a memory or germinal center B cell fate. PI3K-driven effector T cell determination silences TCF1 in one sibling cell, while its PI3K-attenuated sibling self-renews in tandem. Prior to bifurcations achieving irreversible plasma or effector cell fate determination, asymmetric signaling during initial divisions specifies a more proliferative, differentiation-prone lymphocyte in tandem with a more quiescent memory cell sibling. By triggering cell division but transmitting unequal intensity between sibling cells, nutrient-sensitive signaling may be a frequent arbiter of cell fate bifurcations during development and repair.


Assuntos
Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/fisiologia , Células-Tronco Hematopoéticas/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Plasmócitos/citologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/metabolismo , Linhagem da Célula , Citometria de Fluxo , Técnicas de Introdução de Genes , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Plasmócitos/metabolismo , Transdução de Sinais/fisiologia
11.
Immunol Cell Biol ; 93(2): 136-46, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25310967

RESUMO

Signalling through Toll-like receptors (TLRs) by endogenous components of viruses or bacteria can promote antibody (Ab) isotype switching to IgG2a/c. Multiple cell types are capable of responding to TLR stimulation in vivo and the processes underlying TLR-induced Ab isotype switching are not fully defined. Here, we used feeble mice, which are deficient in the peptide/histidine transporter solute carrier family 15 member 4 (Slc15a4), and fail to produce cytokines including interferon alpha (IFNα) in response to TLR9 stimulation, to study Ab isotype switching to IgG2c in response to vaccination. We demonstrate that the production of IgG2c in response to CpGA-adjuvanted vaccines was severely reduced in feeble mice, while a more subtle defect was observed for CpGB. The reduced IgG2c production in feeble could not be ascribed to defective plasmacytoid dendritic cell (pDC) responses alone as we found that splenic cDCs and B cells from feeble mice were also defective in response to TLR9 ligation ex vivo. We conclude that Slc15a4 is required for intact function of TLR9-expressing cells and for effective Ab isotype switching to IgG2c in response to CpG-adjuvanted vaccines.


Assuntos
Switching de Imunoglobulina , Proteínas de Membrana Transportadoras/metabolismo , Receptores de IgG/metabolismo , Recombinação Genética , Receptor Toll-Like 9/metabolismo , Adjuvantes Imunológicos/farmacologia , Animais , Formação de Anticorpos/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Epitopos/imunologia , Imunização , Switching de Imunoglobulina/efeitos dos fármacos , Subpopulações de Linfócitos/efeitos dos fármacos , Subpopulações de Linfócitos/imunologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/farmacologia , Recombinação Genética/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
12.
Nat Rev Immunol ; 14(10): 699-704, 2014 10.
Artigo em Inglês | MEDLINE | ID: mdl-25190286

RESUMO

The cellular progeny of a clonally selected lymphocyte must execute function. However, their function must often occur in more than one way, in more than one place and at more than one time. Experimental evidence supports the view that a single activated lymphocyte can produce a variety of cellular descendants. The mechanisms that are responsible for generating diversity among the progeny of a single lymphocyte remain a subject of lively controversy. Some groups have suggested stochastic mechanisms that are analogous to the diversification of the antigen receptor repertoire. We suggest that the complexity of lymphocyte fates in space and time can be derived from a single naive lymphocyte using the principles of cell diversification that are common in developmental and regenerative biology, including (but not limited to) asymmetric cell division.


Assuntos
Diferenciação Celular/imunologia , Ativação Linfocitária/imunologia , Linfócitos/imunologia , Modelos Imunológicos , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Divisão Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Humanos , Linfócitos/citologia , Linfócitos/metabolismo , Receptores de Antígenos de Linfócitos B/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
13.
J Immunol ; 191(1): 60-9, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23729440

RESUMO

Advances in HIV-1 vaccine clinical trials and preclinical research indicate that the virus envelope glycoproteins (Env) are likely to be an essential component of a prophylactic vaccine. Efficient Ag uptake and presentation by dendritic cells (DCs) is important for strong CD4(+) Th cell responses and the development of effective humoral immune responses. In this study, we examined the capacity of distinct primary human DC subsets to internalize and present recombinant Env to CD4(+) T cells. Consistent with their specific receptor expression, skin DCs bound and internalized Env via C-type lectin receptors, whereas blood DC subsets, including CD1c(+) myeloid DCs, CD123(+) plasmacytoid DCs (PDCs), and CD141(+) DCs exhibited a restricted repertoire of C-type lectin receptors and relied on CD4 for uptake of Env. Despite a generally poor capacity for Ag uptake compared with myeloid DCs, the high expression of CD4 on PDCs allowed them to bind and internalize Env very efficiently. CD4-mediated uptake delivered Env to EEA1(+) endosomes that progressed to Lamp1(+) and MHC class II(+) lysosomes where internalized Env was degraded rapidly. Finally, all three blood DC subsets were able to internalize an Env-CMV pp65 fusion protein via CD4 and stimulate pp65-specific CD4(+) T cells. Thus, in the in vitro systems described in this paper, CD4-mediated uptake of Env is a functional pathway leading to Ag presentation, and this may therefore be a mechanism used by blood DCs, including PDCs, for generating immune responses to Env-based vaccines.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Proteína gp120 do Envelope de HIV/imunologia , HIV-1/imunologia , Linfócitos T CD4-Positivos/metabolismo , Células Dendríticas/metabolismo , Proteína gp120 do Envelope de HIV/sangue , Proteína gp120 do Envelope de HIV/metabolismo , HIV-1/química , HIV-1/metabolismo , Humanos , Lectinas Tipo C/imunologia , Lectinas Tipo C/metabolismo , Ligação Proteica/imunologia , Transporte Proteico/imunologia , Pele/imunologia , Pele/metabolismo , Pele/virologia
14.
J Autoimmun ; 42: 62-70, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23265639

RESUMO

Autoimmune polyendocrine syndrome Type I (APS I) results in multiple endocrine organ destruction and is caused by mutations in the Autoimmune regulator gene (AIRE). In the thymic stroma, cells expressing the AIRE gene dictate T cell education and central tolerance. Although this function is the most studied, AIRE is also expressed in the periphery in DCs and stromal cells. Still, how AIRE regulated transcription modifies cell behaviour in the periphery is largely unknown. Here we show that AIRE is specifically expressed by 33D1(+) DCs and dictates the fate of antibody secreting cell movement within the spleen. We also found that AIRE expressing 33D1(+) DCs expresses self-antigens as exemplified by the hallmark gene insulin. Also, as evidence for a regulatory function, absence of Aire in 33D1(+) DCs led to reduced levels of the chemokine CXCL12 and increased co-stimulatory properties. This resulted in altered activation and recruitment of T-follicular helper cells and germinal centre B cells. The altered balance leads to a change of the early response to a T cell-dependent antigen in Aire(-/-) mice. These findings add to the understanding of how specific DC subtypes regulate the early responses during T cell-dependent antibody responses within the spleen and further define the role of AIRE in the periphery as regulator of self-antigen expression and lymphocyte migration.


Assuntos
Linfócitos B/imunologia , Células Dendríticas Foliculares/imunologia , Poliendocrinopatias Autoimunes/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Fatores de Transcrição/metabolismo , Imunidade Adaptativa/genética , Animais , Formação de Anticorpos/genética , Movimento Celular/genética , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Tolerância Imunológica/genética , Insulina/imunologia , Ativação Linfocitária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Poliendocrinopatias Autoimunes/genética , Fatores de Transcrição/genética , Proteína AIRE
15.
Inhal Toxicol ; 24(7): 401-15, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22642289

RESUMO

CONTEXT: A better understanding of individual subject ozone (O(3)) exposure response kinetics will provide insight into how to improve models used in the risk assessment of ambient ozone exposure. OBJECTIVE: To develop a simple two compartment exposure-response model that describes individual subject decrements in forced expiratory volume in one second (FEV(1)) induced by the acute inhalation of O(3) lasting up to 8 h. METHODS: FEV(1) measurements of 220 subjects who participated in 14 previously completed studies were fit to the model using both particle swarm and nonlinear least squares optimization techniques to identify three subject-specific coefficients producing minimum "global" and local errors, respectively. Observed and predicted decrements in FEV(1) of the 220 subjects were used for validation of the model. Further validation was provided by comparing the observed O(3)-induced FEV(1) decrements in an additional eight studies with predicted values obtained using model coefficients estimated from the 220 subjects used in cross validation. RESULTS: Overall the individual subject measured and modeled FEV(1) decrements were highly correlated (mean R(2) of 0.69 ± 0.24). In addition, it was shown that a matrix of individual subject model coefficients can be used to predict the mean and variance of group decrements in FEV(1). CONCLUSION: This modeling approach provides insight into individual subject O(3) exposure response kinetics and provides a potential starting point for improving the risk assessment of environmental O(3) exposure.


Assuntos
Poluentes Atmosféricos/toxicidade , Volume Expiratório Forçado/efeitos dos fármacos , Modelos Biológicos , Ozônio/toxicidade , Administração por Inalação , Adolescente , Adulto , Ensaios Clínicos como Assunto , Relação Dose-Resposta a Droga , Feminino , Humanos , Exposição por Inalação/efeitos adversos , Cinética , Masculino , Adulto Jovem
16.
J Gen Virol ; 93(Pt 6): 1339-1344, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22357750

RESUMO

The complement-regulatory protein CD46 is the primary receptor for human adenovirus type 35 (HAdV-35) and can regulate human immune-cell activation. CD4(+) T-cells are critical for initiating and maintaining adaptive immunity elicited by infection or vaccination. It was reported previously that HAdV-35 can bind these cells and suppress their activation. The data reported here demonstrate that recombinant trimeric HAdV-35 knob proteins alone can induce CD46 receptor downregulation and inhibit interleukin-2 production and proliferation of human CD4(+) T-cells in vitro similarly to mAbs specific to the CD46 region bound by HAdV-35 knobs. A mutant knob protein with increased affinity for CD46 compared with the wild-type knob caused equivalent effects. In contrast, a CD46-binding-deficient mutant knob protein did not inhibit T-cell activation. Thus, the capacity of HAdV-35 to attenuate human CD4(+) T-cell activation depends predominantly on knob interactions with CD46 and can occur independently of infection.


Assuntos
Infecções por Adenovirus Humanos/imunologia , Adenovírus Humanos/imunologia , Linfócitos T CD4-Positivos/imunologia , Proteínas do Capsídeo/imunologia , Infecções por Adenovirus Humanos/genética , Infecções por Adenovirus Humanos/virologia , Adenovírus Humanos/genética , Linfócitos T CD4-Positivos/virologia , Proteínas do Capsídeo/genética , Células Cultivadas , Regulação para Baixo , Humanos , Interleucina-2/genética , Interleucina-2/imunologia , Ativação Linfocitária , Proteína Cofatora de Membrana/genética , Proteína Cofatora de Membrana/imunologia , Receptores Virais/genética , Receptores Virais/imunologia
17.
Methods Mol Biol ; 820: 215-30, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22131034

RESUMO

Type-1 interferons (IFNs), including IFNα/ß, are a family of cytokines produced rapidly upon pathogen encounter and crucial for bridging innate and adaptive immunity. IFNα has been widely appreciated as a multifunctional cytokine involved particularly in early immune responses against viral, bacterial, and parasitic infections. Although most cells may be competent to produce IFNα during specific conditions, plasmacytoid dendritic cells (PDCs) are unique in their capacity to produce rapid and robust levels in response to various pathogens. PDCs to a great extent utilize toll-like receptor (TLR) 7 and 9, localized in early endosomes, to sense pathogen-associated nucleic acids, and initiate the signaling cascade leading to induction of IFNα. Here, we provide basic protocols for the detection of IFNα in individual immune cells, particularly PDCs, using flow cytometry. We discuss the key elements for successful isolation of PDCs, stimulation, immunostaining, and identification of IFNα producing cells.


Assuntos
Células Dendríticas/metabolismo , Interferon-alfa/isolamento & purificação , Interferon-alfa/metabolismo , Células Cultivadas , Células Dendríticas/imunologia , Citometria de Fluxo , Humanos , Fenômenos do Sistema Imunitário , Imunidade Inata/imunologia , Transdução de Sinais/imunologia , Receptor 7 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo
18.
J Invest Dermatol ; 132(1): 114-23, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21850028

RESUMO

Plasmacytoid dendritic cells (pDCs) are rarely present in normal skin but have been shown to infiltrate lesions of infections or autoimmune disorders. Here, we report that several DC subsets including CD123(+) BDCA-2/CD303(+) pDCs accumulate in the dermis in indurations induced by the tuberculin skin test (TST), used to screen immune sensitization by Mycobacterium tuberculosis. Although the purified protein derivate (PPD) used in the TST did not itself induce pDC recruitment or IFN-α production, the positive skin reactions showed high expression of the IFN-α-inducible protein MxA. In contrast, the local immune response to PPD was associated with substantial cell death and high expression of the cationic antimicrobial peptide LL37, which together can provide a means for pDC activation and IFN-α production. In vitro, pDCs showed low uptake of PPD compared with CD11c(+) and BDCA-3/CD141(+) myeloid DC subsets. Furthermore, supernatants from pDCs activated with LL37-DNA complexes reduced the high PPD uptake in myeloid DCs, as well as decreased their capacity to activate T-cell proliferation. Infiltrating pDCs in the TST reaction site may thus have a regulatory effect upon the antigen processing and presentation functions of surrounding potent myeloid DC subsets to limit potentially detrimental and excessive immune stimulation.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Mycobacterium tuberculosis/imunologia , Teste Tuberculínico , Tuberculose/imunologia , Peptídeos Catiônicos Antimicrobianos , Biomarcadores/metabolismo , Biópsia , Catelicidinas/genética , Catelicidinas/imunologia , Comunicação Celular/imunologia , Morte Celular/imunologia , Movimento Celular/imunologia , Células Dendríticas/patologia , Proteínas de Ligação ao GTP/metabolismo , Humanos , Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Proteínas de Resistência a Myxovirus , Pele/imunologia , Pele/microbiologia , Pele/patologia , Linfócitos T/imunologia , Linfócitos T/patologia , Tuberculose/diagnóstico
19.
Proc Natl Acad Sci U S A ; 108(18): 7499-504, 2011 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-21502499

RESUMO

Recombinant adenoviruses (rAds) based on types 5 (rAd5) and 35 (rAd35) have emerged as important vaccine delivery vectors in clinical testing for a variety of pathogens. A major difference between these vectors is their binding to cellular receptors used for infection. Whereas rAd5 binds coxsackie-adenovirus receptor (CAR), rAd35 binds the complement regulatory protein CD46. Although rAd35 infected and phenotypically matured human blood dendritic cells (DCs) more efficiently than rAd5, we show here that rAd35 markedly suppressed DC-induced activation of naive CD4(+) T cells. rAd35 specifically blocked both DCs and anti-CD3/CD28 mAb-induced naive T-cell proliferation and IL-2 production. This effect was also observed in CD4(+) memory T cells but to a lesser extent. The suppression occurred by rAd35 binding to CD46 on T cells and was independent of infection. CD46 engagement with mAb mimicked the effects of rAd35 and also led to deficient NF-κB nuclear translocation. In contrast, rAd5 and rAd35 vectors with ablated CD46 binding did not inhibit T-cell activation. Our findings provide insights into the basic biology of adenoviruses and indicate that CD46 binding may have an impact on the generation of primary CD4(+) T-cell responses by Ad35.


Assuntos
Adenoviridae/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Vetores Genéticos/metabolismo , Ativação Linfocitária/fisiologia , Proteína Cofatora de Membrana/metabolismo , Receptores Virais/metabolismo , Adenoviridae/genética , Western Blotting , Linfócitos T CD4-Positivos/imunologia , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Células Dendríticas/metabolismo , Citometria de Fluxo , Humanos
20.
J Leukoc Biol ; 89(6): 811-21, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21233412

RESUMO

The development and quality of a humoral immune response are largely influenced by the environment that supports the activation of naïve B cells. Human PDCs, through their unique capacity to produce high levels of IFN-α, have been shown earlier to enhance B cell responses stimulated by selected TLR ligands. In this study, we investigated whether PDCs also promote B cell activation induced by Th cell interactions and BCR ligation. Sorted human naive CD19(+) CD27(-) B cells were activated in vitro with anti-Ig and irradiated CD4(+) T cells. Under these conditions, the presence of supernatants from TLR-stimulated PDCs increased B cell proliferation, the frequency of B cells that differentiated to CD27(high) CD38(high) cells, and secretion of IgM. Similar results were observed when the B cells were activated in the presence of purified IFN-α. In contrast, supernatants from stimulated MDCs did not augment these functions. Also, IFN-α treatment of B cells up-regulated the expression of costimulatory molecule CD86 but not CD40, CD80, MHC class II, or CD25. Although direct IFN-α exposure of T cells suppressed their proliferative capacity, IFN-α treatment of B cells led to a small increase in their capacity to induce superantigen-driven activation of autologous CD4(+) T cells. In summary, PDCs, via their production of IFN-α, may render B cells more responsive to T cell contact, which in turn, facilitates B cell proliferation and differentiation to antibody-producing cells.


Assuntos
Células Produtoras de Anticorpos/imunologia , Linfócitos B/imunologia , Diferenciação Celular/imunologia , Células Dendríticas/metabolismo , Interferon-alfa/metabolismo , Linfócitos T/imunologia , Células Cultivadas , Citocinas/metabolismo , Humanos , Imunoglobulina M/metabolismo , Memória Imunológica , Ativação Linfocitária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA