Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Sci Transl Med ; 13(623): eabh1962, 2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34878825

RESUMO

Despite the high remission rates achieved using T cells bearing a chimeric antigen receptor (CAR) against hematogical malignancies, there is still a considerable proportion of patients who eventually experience tumor relapse. Clinical studies have established that mechanisms of treatment failure include the down-regulation of target antigen expression and the limited persistence of effective CAR T cells. We hypothesized that dual targeting mediated by a CAR and a chimeric costimulatory receptor (CCR) could simultaneously enhance T cell cytotoxicity and improve durability. Concomitant high-affinity engagement of a CD38-binding CCR enhanced the cytotoxicity of BCMA-CAR and CD19-CAR T cells by increasing their functional binding avidity. In comparison to second-generation BCMA-CAR or CD19-CAR T cells, double-targeted CAR + CD38-CCR T cells exhibited increased sensitivity to recognize and lyse tumor variants of multiple myeloma and acute lymphoblastic leukemia with low antigen density in vitro. In addition, complimentary costimulation by 4-1BB and CD28 endodomains provided by the CAR and CCR combination conferred increased cytokine secretion and expansion and improved persistence in vivo. The cumulatively improved properties of CAR + CCR T cells enabled the in vivo eradication of antigen-low tumor clones, which were otherwise resistant to treatment with conventional CAR T cells. Therefore, multiplexing targeting and costimulation through the combination of a CAR and a CCR is a powerful strategy to improve the clinical outcomes of CAR T cells by enhancing cytotoxic efficacy and persistence, thus preventing relapses of tumor clones with low target antigen density.


Assuntos
Mieloma Múltiplo , Receptores de Antígenos Quiméricos , Antígenos CD19 , Humanos , Imunoterapia Adotiva , Mieloma Múltiplo/terapia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T
2.
Mucosal Immunol ; 13(1): 140-148, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31636345

RESUMO

Immune responses following Mycobacterium tuberculosis (Mtb) infection or vaccination are frequently assessed by measuring T-cell recognition of crude Mtb antigens, recombinant proteins, or peptide epitopes. We previously showed that not all Mtb-specific T cells recognize Mtb-infected macrophages. Thus, an important question is what proportion of T cells elicited by Mtb infection recognize Mtb-infected macrophages. We address this question by developing a modified elispot assay using viable Mtb-infected macrophages, a low multiplicity of infection and purified T cells. In C57BL/6 mice, CD4 and CD8 T cells were classically MHC restricted. Comparable frequencies of T cells that recognize Mtb-infected macrophages were determined using interferon-γ elispot and intracellular cytokine staining, and lung CD4 T cells more sensitively recognized Mtb-infected macrophages than lung CD8 T cells. Compared to the relatively high frequencies of T cells specific for antigens such as ESAT-6 and TB10.4, low frequencies of total pulmonary T cells elicited by aerosolized Mtb infection recognize Mtb-infected macrophages. Finally, we demonstrate that BCG vaccination elicits T cells that recognize Mtb-infected macrophages. We propose that the frequency of T cells that recognize infected macrophages could correlate with protective immunity and may be an alternative approach to measuring T-cell responses to Mtb antigens.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Pulmão/imunologia , Macrófagos/imunologia , Mycobacterium tuberculosis/fisiologia , Tuberculose/imunologia , Animais , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Células Cultivadas , ELISPOT , Humanos , Interferon gama/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium bovis/imunologia , Vacinação
3.
Am J Physiol Lung Cell Mol Physiol ; 314(2): L225-L235, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28982735

RESUMO

Acidic microenvironments commonly occur at sites of inflammation and bacterial infections. In the context of a Pseudomonas aeruginosa infection, we previously demonstrated that acidosis enhances the cellular proinflammatory interleukin (IL)-1ß response in vitro. However, how pH alterations affect in vivo IL-1ß responses and subsequent IL-1-driven inflammation during infection with P. aeruginosa is unclear. Here, we report that acidosis enhances in vivo IL-1ß production and downstream IL-1 receptor-dependent responses during infection with P. aeruginosa in models of acute pneumonia and peritonitis. Importantly, we demonstrate that infection with P. aeruginosa within an acidic environment leads to enhanced production of a subset of proinflammatory cytokines, including chemokine (C-X-C) motif ligand 1, IL-6, and chemokine (C-C motif) ligand 2, and increased neutrophil recruitment. Furthermore, with the use of IL-1 receptor type 1-deficient mice, we identify the contribution of the IL-1 signaling pathway to the acidosis-enhanced inflammatory response and pathology. These data provide insights into the potential benefit of pH regulation during bacterial infections to control disease progression and immunopathology.


Assuntos
Acidose/fisiopatologia , Inflamação/imunologia , Interleucina-1/farmacologia , Pneumonia Bacteriana/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Receptores Tipo I de Interleucina-1/fisiologia , Animais , Citocinas/metabolismo , Feminino , Inflamação/microbiologia , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia
4.
Toxicol Sci ; 159(1): 64-75, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28903492

RESUMO

Complex interactions between genetic and environmental factors are widely believed to underlie the incidence and progression of Parkinson's disease (PD). Rotenone is a naturally occurring metabolic toxin employed as an insecticide and piscicide identified as a risk factor for the development of PD in agricultural workers. The Nlrp3 inflammasome is an intracellular mediator that can initiate an inflammatory cascade in response to cellular stress. Reports by others indicating that NLRP3 expression was detectable in tissues obtained from Alzheimer's disease patients and that the PD-associated protein α-synuclein could activate inflammasomes in cultured glial cells, prompted us to test the prediction that Nlrp3 was required for the development of Parkinson's-like changes resulting from rotenone exposure in mice. We exposed wild type and Nlrp3-/- mice to chronic low doses of intragastric rotenone and conducted longitudinal behavioral and serum cytokine analysis followed by evaluation of neuroinflammatory and neurodegenerative endpoints in brain tissues. We observed progressive rotenone-dependent changes in serum cytokine levels and circulating leukocytes in wild type mice not observed in Nlrp3-/- mice. Analysis of brain tissues revealed Nlrp3-dependent neuroinflammation and nigral cell loss in mice exposed to rotenone as compared with mice exposed to vehicle alone. Together, our findings provide compelling evidence of a role for Nlrp3 in nigral degeneration and neuroinflammation resulting from systemic rotenone exposure and suggest that the suppression of NLRP3 activity may be a rational neuroprotective strategy for toxin-associated PD.


Assuntos
Inflamação/fisiopatologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Rotenona/toxicidade , Estômago , Substância Negra/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Estresse Oxidativo , Doença de Parkinson/fisiopatologia , Rotenona/administração & dosagem , Substância Negra/patologia , Testes de Toxicidade Crônica
5.
Am J Physiol Lung Cell Mol Physiol ; 309(8): L902-13, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26472815

RESUMO

The NLRC4 inflammasome is responsible for IL-1ß processing by macrophages in response to Pseudomonas aeruginosa infection. We therefore hypothesized that mice that lack ASC, an NLRC4 inflammasome adaptor protein necessary for in vitro IL-1ß production by macrophages, would be preferentially protected from a hyperinflammatory lethal challenge that is dependent on bacterial type three secretion system (T3SS) activity. We report herein that lack of ASC does not confer preferential protection in response to P. aeruginosa acute infection and that ASC(-/-) mice are capable of producing robust amounts of IL-1ß comparable with C57BL/6 mice. We now identify that neutrophils represent the ASC-independent source of IL-1ß production during the acute phases of infection both in models of acute pneumonia and peritonitis. Consequently, depletion of neutrophils in ASC(-/-) mice leads to a marked deficit in IL-1ß production in vivo. The pulmonary neutrophil IL-1ß response is predominantly dependent on caspase-1, which contrasts with data derived from ocular infection. These studies therefore identify a noncanonical mechanism of IL-1ß production by neutrophils independent of ASC and demonstrate the first physiological contribution of neutrophils as an important source of IL-1ß in response to acute P. aeruginosa infection during acute pneumonia and peritonitis.


Assuntos
Proteínas Reguladoras de Apoptose/imunologia , Interleucina-1beta/biossíntese , Neutrófilos/imunologia , Neutrófilos/microbiologia , Pseudomonas aeruginosa/imunologia , Pseudomonas aeruginosa/patogenicidade , Animais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Ligação ao Cálcio/deficiência , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/imunologia , Modelos Animais de Doenças , Técnicas In Vitro , Inflamassomos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peritonite/imunologia , Pneumonia Bacteriana/imunologia , Infecções por Pseudomonas/imunologia
6.
Infect Immun ; 82(11): 4689-97, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25156732

RESUMO

Infection by Pseudomonas aeruginosa, and bacteria in general, frequently promotes acidification of the local microenvironment, and this is reinforced by pulmonary exertion and exacerbation. However, the consequence of an acidic environment on the host inflammatory response to P. aeruginosa infection is poorly understood. Here we report that the pivotal cellular and host proinflammatory interleukin-1ß (IL-1ß) response, which enables host clearance of the infection but can produce collateral inflammatory damage, is increased in response to P. aeruginosa infection within an acidic environment. Synergistic mechanisms that promote increased IL-1ß release in response to P. aeruginosa infection in an acidic environment are increased pro-IL-1ß induction and increased caspase-1 activity, the latter being dependent upon a functional type III secretion system of the bacteria and the NLRC4 inflammasome of the host. Using an in vivo peritonitis model, we have validated that the IL-1ß inflammatory response is increased in mice in response to P. aeruginosa infection within an acidic microenvironment. These data reveal novel insights into the regulation and exacerbation of inflammatory responses to P. aeruginosa.


Assuntos
Acidose/metabolismo , Regulação da Expressão Gênica/imunologia , Interleucina-1beta/metabolismo , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/fisiologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspase 1/metabolismo , Concentração de Íons de Hidrogênio , Imunidade Inata , Interleucina-1beta/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/metabolismo
7.
Am J Physiol Lung Cell Mol Physiol ; 306(7): L591-603, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24464809

RESUMO

Pseudomonas aeruginosa is an opportunistic bacterial pathogen responsible for a high incidence of acute and chronic pulmonary infection. These infections are particularly prevalent in patients with chronic obstructive pulmonary disease and cystic fibrosis: much of the morbidity and pathophysiology associated with these diseases is due to a hypersusceptibility to bacterial infection. Innate immunity, primarily through inflammatory cytokine production, cellular recruitment, and phagocytic clearance by neutrophils and macrophages, is the key to endogenous control of P. aeruginosa infection. In this review, we highlight recent advances toward understanding the innate immune response to P. aeruginosa, with a focus on the role of phagocytes in control of P. aeruginosa infection. Specifically, we summarize the cellular and molecular mechanisms of phagocytic recognition and uptake of P. aeruginosa, and how current animal models of P. aeruginosa infection reflect clinical observations in the context of phagocytic clearance of the bacteria. Several notable phenotypic changes to the bacteria are consistently observed during chronic pulmonary infections, including changes to mucoidy and flagellar motility, that likely enable or reflect their ability to persist. These traits are likewise examined in the context of how the bacteria avoid phagocytic clearance, inflammation, and sterilizing immunity.


Assuntos
Fagocitose/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Infecções Respiratórias/imunologia , Animais , Proteínas de Bactérias/metabolismo , Flagelos/fisiologia , Humanos , Imunidade Inata , Inflamassomos/fisiologia , Inflamação/microbiologia , Pulmão/imunologia , Pulmão/microbiologia , Macrófagos/imunologia , Metaloendopeptidases/metabolismo , Modelos Animais , Neutrófilos/imunologia , Polissacarídeos Bacterianos/metabolismo , Proteína C/fisiologia , Pseudomonas aeruginosa/patogenicidade , Pseudomonas aeruginosa/fisiologia , Receptores Imunológicos/fisiologia , Infecções Respiratórias/microbiologia
8.
Infect Immun ; 81(6): 2043-52, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23529619

RESUMO

We previously demonstrated that bacterial flagellar motility is a fundamental mechanism by which host phagocytes bind and ingest bacteria. Correspondingly, loss of bacterial motility, consistently observed in clinical isolates from chronic Pseudomonas aeruginosa infections, enables bacteria to evade association and ingestion of P. aeruginosa by phagocytes both in vitro and in vivo. Since bacterial interactions with the phagocyte cell surface are required for type three secretion system-dependent NLRC4 inflammasome activation by P. aeruginosa, we hypothesized that reduced bacterial association with phagocytes due to loss of bacterial motility, independent of flagellar expression, will lead to reduced inflammasome activation. Here we report that inflammasome activation is reduced in response to nonmotile P. aeruginosa. Nonmotile P. aeruginosa elicits reduced IL-1ß production as well as caspase-1 activation by peritoneal macrophages and bone marrow-derived dendritic cells in vitro. Importantly, nonmotile P. aeruginosa also elicits reduced IL-1ß levels in vivo in comparison to those elicited by wild-type P. aeruginosa. This is the first demonstration that loss of bacterial motility results in reduced inflammasome activation and antibacterial IL-1ß host response. These results provide a critical insight into how the innate immune system responds to bacterial motility and, correspondingly, how pathogens have evolved mechanisms to evade the innate immune system.


Assuntos
Flagelos/fisiologia , Inflamassomos/metabolismo , Movimento/fisiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Proteínas Adaptadoras de Sinalização CARD , Caspase 1/genética , Caspase 1/metabolismo , Morte Celular , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Células Dendríticas/metabolismo , Regulação da Expressão Gênica/imunologia , Inflamassomos/genética , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/citologia , Pseudomonas aeruginosa/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA