Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 602(7895): 156-161, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34847567

RESUMO

CD8 T cell-mediated autoimmune diseases result from the breakdown of self-tolerance mechanisms in autoreactive CD8 T cells1. How autoimmune T cell populations arise and are sustained, and the molecular programmes defining the autoimmune T cell state, are unknown. In type 1 diabetes, ß-cell-specific CD8 T cells destroy insulin-producing ß-cells. Here we followed the fate of ß-cell-specific CD8 T cells in non-obese diabetic mice throughout the course of type 1 diabetes. We identified a stem-like autoimmune progenitor population in the pancreatic draining lymph node (pLN), which self-renews and gives rise to pLN autoimmune mediators. pLN autoimmune mediators migrate to the pancreas, where they differentiate further and destroy ß-cells. Whereas transplantation of as few as 20 autoimmune progenitors induced type 1 diabetes, as many as 100,000 pancreatic autoimmune mediators did not. Pancreatic autoimmune mediators are short-lived, and stem-like autoimmune progenitors must continuously seed the pancreas to sustain ß-cell destruction. Single-cell RNA sequencing and clonal analysis revealed that autoimmune CD8 T cells represent unique T cell differentiation states and identified features driving the transition from autoimmune progenitor to autoimmune mediator. Strategies aimed at targeting the stem-like autoimmune progenitor pool could emerge as novel and powerful immunotherapeutic interventions for type 1 diabetes.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Células Secretoras de Insulina/imunologia , Células-Tronco/patologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/transplante , Autorrenovação Celular , Células Clonais/imunologia , Células Clonais/metabolismo , Células Clonais/patologia , Modelos Animais de Doenças , Feminino , Glucose-6-Fosfatase/imunologia , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Células Secretoras de Insulina/patologia , Linfonodos/imunologia , Masculino , Camundongos , Receptores de Antígenos de Linfócitos T/metabolismo , Análise de Célula Única , Transplante de Células-Tronco , Células-Tronco/imunologia , Células-Tronco/metabolismo , Transcriptoma
2.
Proc Natl Acad Sci U S A ; 120(3): e2205049120, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36634134

RESUMO

Stimulator of interferon genes (STING) signaling has been extensively studied in inflammatory diseases and cancer, while its role in T cell responses to infection is unclear. Using Listeria monocytogenes strains engineered to induce different levels of c-di-AMP, we found that high STING signals impaired T cell memory upon infection via increased Bim levels and apoptosis. Unexpectedly, reduction of TCR signal strength or T cell-STING expression decreased Bim expression, T cell apoptosis, and recovered T cell memory. We found that TCR signal intensity coupled STING signal strength to the unfolded protein response (UPR) and T cell survival. Under strong STING signaling, Indoleamine-pyrrole 2,3-dioxygenase (IDO) inhibition also reduced apoptosis and led to a recovery of T cell memory in STING sufficient CD8 T cells. Thus, STING signaling regulates CD8 T cell memory fitness through both cell-intrinsic and extrinsic mechanisms. These studies provide insight into how IDO and STING therapies could improve long-term T cell protective immunity.


Assuntos
Receptores de Antígenos de Linfócitos T , Transdução de Sinais , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T CD8-Positivos , Células T de Memória , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo
3.
Immunity ; 45(2): 389-401, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27521269

RESUMO

CD8(+) T cells recognizing tumor-specific antigens are detected in cancer patients but are dysfunctional. Here we developed a tamoxifen-inducible liver cancer mouse model with a defined oncogenic driver antigen (SV40 large T-antigen) to follow the activation and differentiation of naive tumor-specific CD8(+) T (TST) cells after tumor initiation. Early during the pre-malignant phase of tumorigenesis, TST cells became dysfunctional, exhibiting phenotypic, functional, and transcriptional features similar to dysfunctional T cells isolated from late-stage human tumors. Thus, T cell dysfunction seen in advanced human cancers may already be established early during tumorigenesis. Although the TST cell dysfunctional state was initially therapeutically reversible, it ultimately evolved into a fixed state. Persistent antigen exposure rather than factors associated with the tumor microenvironment drove dysfunction. Moreover, the TST cell differentiation and dysfunction program exhibited features distinct from T cell exhaustion in chronic infections. Strategies to overcome this antigen-driven, cell-intrinsic dysfunction may be required to improve cancer immunotherapy.


Assuntos
Antígenos Transformantes de Poliomavirus/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Imunoterapia Adotiva/métodos , Neoplasias Hepáticas/imunologia , Animais , Carcinogênese , Diferenciação Celular , Células Cultivadas , Senescência Celular , Modelos Animais de Doenças , Humanos , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/terapia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tamoxifeno , Microambiente Tumoral
4.
Nature ; 571(7764): 270-274, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31207604

RESUMO

Tumour-specific CD8 T cell dysfunction is a differentiation state that is distinct from the functional effector or memory T cell states1-6. Here we identify the nuclear factor TOX as a crucial regulator of the differentiation of tumour-specific T (TST) cells. We show that TOX is highly expressed in dysfunctional TST cells from tumours and in exhausted T cells during chronic viral infection. Expression of TOX is driven by chronic T cell receptor stimulation and NFAT activation. Ectopic expression of TOX in effector T cells in vitro induced a transcriptional program associated with T cell exhaustion. Conversely, deletion of Tox in TST cells in tumours abrogated the exhaustion program: Tox-deleted TST cells did not upregulate genes for inhibitory receptors (such as Pdcd1, Entpd1, Havcr2, Cd244 and Tigit), the chromatin of which remained largely inaccessible, and retained high expression of transcription factors such as TCF-1. Despite their normal, 'non-exhausted' immunophenotype, Tox-deleted TST cells remained dysfunctional, which suggests that the regulation of expression of inhibitory receptors is uncoupled from the loss of effector function. Notably, although Tox-deleted CD8 T cells differentiated normally to effector and memory states in response to acute infection, Tox-deleted TST cells failed to persist in tumours. We hypothesize that the TOX-induced exhaustion program serves to prevent the overstimulation of T cells and activation-induced cell death in settings of chronic antigen stimulation such as cancer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Diferenciação Celular/imunologia , Proteínas de Grupo de Alta Mobilidade/metabolismo , Proteínas de Homeodomínio/metabolismo , Neoplasias/imunologia , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Grupo de Alta Mobilidade/deficiência , Proteínas de Grupo de Alta Mobilidade/genética , Proteínas de Homeodomínio/genética , Humanos , Memória Imunológica , Linfócitos do Interstício Tumoral/citologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/patologia , Camundongos , Neoplasias/patologia , Fenótipo , Receptores de Antígenos de Linfócitos T/imunologia , Transcrição Gênica
5.
Nature ; 545(7655): 452-456, 2017 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-28514453

RESUMO

Tumour-specific CD8 T cells in solid tumours are dysfunctional, allowing tumours to progress. The epigenetic regulation of T cell dysfunction and therapeutic reprogrammability (for example, to immune checkpoint blockade) is not well understood. Here we show that T cells in mouse tumours differentiate through two discrete chromatin states: a plastic dysfunctional state from which T cells can be rescued, and a fixed dysfunctional state in which the cells are resistant to reprogramming. We identified surface markers associated with each chromatin state that distinguished reprogrammable from non-reprogrammable PD1hi dysfunctional T cells within heterogeneous T cell populations from tumours in mice; these surface markers were also expressed on human PD1hi tumour-infiltrating CD8 T cells. Our study has important implications for cancer immunotherapy as we define key transcription factors and epigenetic programs underlying T cell dysfunction and surface markers that predict therapeutic reprogrammability.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Cromatina/genética , Cromatina/metabolismo , Neoplasias/genética , Neoplasias/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Epigênese Genética , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Memória Imunológica , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/patologia , Masculino , Camundongos , Neoplasias/metabolismo , Neoplasias/terapia , Fatores de Transcrição/metabolismo
6.
Nature ; 517(7533): 170-3, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25567281

RESUMO

Intracellular pathogens are responsible for much of the world-wide morbidity and mortality due to infectious diseases. To colonize their hosts successfully, pathogens must sense their environment and regulate virulence gene expression appropriately. Accordingly, on entry into mammalian cells, the facultative intracellular bacterial pathogen Listeria monocytogenes remodels its transcriptional program by activating the master virulence regulator PrfA. Here we show that bacterial and host-derived glutathione are required to activate PrfA. In this study a genetic selection led to the identification of a bacterial mutant in glutathione synthase that exhibited reduced virulence gene expression and was attenuated 150-fold in mice. Genome sequencing of suppressor mutants that arose spontaneously in vivo revealed a single nucleotide change in prfA that locks the protein in the active conformation (PrfA*) and completely bypassed the requirement for glutathione during infection. Biochemical and genetic studies support a model in which glutathione-dependent PrfA activation is mediated by allosteric binding of glutathione to PrfA. Whereas glutathione and other low-molecular-weight thiols have important roles in redox homeostasis in all forms of life, here we demonstrate that glutathione represents a critical signalling molecule that activates the virulence of an intracellular pathogen.


Assuntos
Regulação Bacteriana da Expressão Gênica/genética , Glutationa/metabolismo , Espaço Intracelular/metabolismo , Espaço Intracelular/microbiologia , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidade , Regulação Alostérica/efeitos dos fármacos , Proteínas de Bactérias/metabolismo , DNA/metabolismo , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Glutationa/farmacologia , Espaço Intracelular/efeitos dos fármacos , Listeria monocytogenes/efeitos dos fármacos , Macrófagos/metabolismo , Mutação/genética , Fatores de Terminação de Peptídeos/metabolismo , Ligação Proteica , Seleção Genética/genética , Supressão Genética/genética , Virulência/genética
7.
Proc Natl Acad Sci U S A ; 115(32): 8179-8184, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30038013

RESUMO

Agents that remodel the tumor microenvironment (TME), prime functional tumor-specific T cells, and block inhibitory signaling pathways are essential components of effective immunotherapy. We are evaluating live-attenuated, double-deleted Listeria monocytogenes expressing tumor antigens (LADD-Ag) in the clinic. Here we show in numerous mouse models that while treatment with nonrecombinant LADD induced some changes in the TME, no antitumor efficacy was observed, even when combined with immune checkpoint blockade. In contrast, LADD-Ag promoted tumor rejection by priming tumor-specific KLRG1+PD1loCD62L- CD8+ T cells. These IFNγ-producing effector CD8+ T cells infiltrated the tumor and converted the tumor from an immunosuppressive to an inflamed microenvironment that was characterized by a decrease in regulatory T cells (Treg) levels, a proinflammatory cytokine milieu, and the shift of M2 macrophages to an inducible nitric oxide synthase (iNOS)+CD206- M1 phenotype. Remarkably, these LADD-Ag-induced tumor-specific T cells persisted for more than 2 months after primary tumor challenge and rapidly controlled secondary tumor challenge. Our results indicate that the striking antitumor efficacy observed in mice with LADD-based immunotherapy stems from TME remodeling which is a direct consequence of eliciting potent, systemic tumor-specific CD8+ T cells.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Listeria monocytogenes/imunologia , Neoplasias/terapia , Microambiente Tumoral/imunologia , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/uso terapêutico , Vacinas Anticâncer/genética , Vacinas Anticâncer/uso terapêutico , Linhagem Celular Tumoral , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Listeria monocytogenes/genética , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias/imunologia , Resultado do Tratamento , Vacinação/métodos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/uso terapêutico , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Vacinas de DNA/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Infect Immun ; 87(8)2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31235641

RESUMO

Live-attenuated Listeria monocytogenes has shown encouraging potential as an immunotherapy platform in preclinical and clinical settings. However, additional safety measures will enable application across malignant and infectious diseases. Here, we describe a new vaccine platform, termed Lm-RIID (L. monocytogenes recombinase-induced intracellular death), that induces the deletion of genes required for bacterial viability yet maintains potent T cell responses to encoded antigens. Lm-RIID grows normally in broth but commits suicide inside host cells by inducing Cre recombinase and deleting essential genes flanked by loxP sites, resulting in a self-limiting infection even in immunocompromised mice. Lm-RIID vaccination of mice induces potent CD8+ T cells and protects against virulent challenges, similar to live L. monocytogenes vaccines. When combined with α-PD-1, Lm-RIID is as effective as live-attenuated L. monocytogenes in a therapeutic tumor model. This impressive efficacy, together with the increased clearance rate, makes Lm-RIID ideal for prophylactic immunization against diseases that require T cells for protection.


Assuntos
Vacinas Bacterianas/imunologia , Listeria monocytogenes/imunologia , Animais , Feminino , Imunoterapia , Listeria monocytogenes/patogenicidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia , Vacinas Atenuadas/imunologia , Virulência
9.
Infect Immun ; 86(3)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29263107

RESUMO

Listeria monocytogenes is a Gram-positive intracellular pathogen that causes a severe invasive disease. Upon infecting a host cell, L. monocytogenes upregulates the transcription of numerous factors necessary for productive infection. VirR is the response regulator component of a two-component regulatory system in L. monocytogenes In this report, we have identified the putative ABC transporter encoded by genes lmo1746-lmo1747 as necessary for VirR function. We have designated lmo1746-lmo1747 virAB We constructed an in-frame deletion of virAB and determined that the ΔvirAB mutant exhibited reduced transcription of VirR-regulated genes. The ΔvirAB mutant also showed defects in in vitro plaque formation and in vivo virulence that were similar to those of a ΔvirR deletion mutant. Since VirR is important for innate resistance to antimicrobial agents, we determined the MICs of nisin and bacitracin for ΔvirAB bacteria. We found that VirAB expression was necessary for nisin resistance but was dispensable for resistance to bacitracin. This result suggested a VirAB-independent mechanism of VirR regulation in response to bacitracin. Lastly, we found that the ΔvirR and ΔvirAB mutants had no deficiency in growth in broth culture, intracellular replication, or production of the ActA surface protein, which facilitates actin-based motility and cell-to-cell spread. However, the ΔvirR and ΔvirAB mutants produced shorter actin tails during intracellular infection, which suggested that these mutants have a reduced ability to move and spread via actin-based motility. These findings have demonstrated that L. monocytogenes VirAB functions in a pathway with VirR to regulate the expression of genes necessary for virulence and resistance to antimicrobial agents.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Antibacterianos/farmacologia , Proteínas de Bactérias/metabolismo , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidade , Listeriose/microbiologia , Nisina/farmacologia , Fatores de Transcrição/genética , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Proteínas de Bactérias/genética , Farmacorresistência Bacteriana , Feminino , Regulação Bacteriana da Expressão Gênica , Humanos , Listeria monocytogenes/efeitos dos fármacos , Listeria monocytogenes/genética , Camundongos Endogâmicos BALB C , Testes de Sensibilidade Microbiana , Regulon , Fatores de Transcrição/metabolismo , Virulência
10.
Gastroenterology ; 146(7): 1784-94.e6, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24607504

RESUMO

BACKGROUND & AIMS: Premalignant lesions and early stage tumors contain immunosuppressive microenvironments that create barriers for cancer vaccines. Kras(G12D/+);Trp53(R172H/+);Pdx-1-Cre (KPC) mice, which express an activated form of Kras in pancreatic tissues, develop pancreatic intraepithelial neoplasms (PanIN) that progress to pancreatic ductal adenocarcinoma (PDA). We used these mice to study immune suppression in PDA. METHODS: We immunized KPC and Kras(G12D/+);Pdx-1-Cre mice with attenuated intracellular Listeria monocytogenes (which induces CD4(+) and CD8(+) T-cell immunity) engineered to express Kras(G12D) (LM-Kras). The vaccine was given alone or in sequence with an anti-CD25 antibody (PC61) and cyclophosphamide to deplete T-regulatory (Treg) cells. Survival times were measured; pancreatic and spleen tissues were collected and analyzed by histologic, flow cytometry, and immunohistochemical analyses. RESULTS: Interferon γ-mediated, CD8(+) T-cell responses were observed in KPC and Kras(G12D/+);Pdx-1-Cre mice given LM-Kras, but not in unvaccinated mice. Administration of LM-Kras to KPC mice 4-6 weeks old (with early stage PanINs), depleted of Treg cells, significantly prolonged survival and reduced PanIN progression (median survival, 265 days), compared with unvaccinated mice (median survival, 150 days; P = .002), mice given only LM-Kras (median survival, 150 days; P = .050), and unvaccinated mice depleted of Treg cells (median survival, 170 days; P = .048). In 8- to 12-week-old mice (with late-stage PanINs), LM-Kras, alone or in combination with Treg cell depletion, did not increase survival time or slow PanIN progression. The combination of LM-Kras and Treg cell depletion reduced numbers of Foxp3(+)CD4(+) T cells in pancreatic lymph nodes, increased numbers of CD4(+) T cells that secrete interleukin 17 and interferon γ, and caused CD11b(+)Gr1(+) cells in the pancreas to acquire an immunostimulatory phenotype. CONCLUSIONS: Immunization of KPC mice with Listeria monocytogenes engineered to express Kras(G12D), along with depletion of Treg cells, reduces progression of early stage, but not late-stage, PanINs. This approach increases infiltration of the lesion with inflammatory cells. It might be possible to design immunotherapies against premalignant pancreatic lesions to slow or prevent progression to PDA.


Assuntos
Vacinas Anticâncer/uso terapêutico , Carcinoma in Situ/tratamento farmacológico , Carcinoma Ductal Pancreático/tratamento farmacológico , Listeria monocytogenes/imunologia , Neoplasias Pancreáticas/tratamento farmacológico , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Antígeno CD11b/metabolismo , Vacinas Anticâncer/imunologia , Carcinoma in Situ/genética , Carcinoma in Situ/imunologia , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patologia , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Ciclofosfamida/farmacologia , Modelos Animais de Doenças , Progressão da Doença , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Integrases/genética , Integrases/metabolismo , Interferon gama/metabolismo , Interleucina-17/metabolismo , Listeria monocytogenes/genética , Listeria monocytogenes/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptores de Quimiocinas/metabolismo , Linfócitos T Reguladores/metabolismo , Fatores de Tempo , Transativadores/genética , Transativadores/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
11.
J Immunol ; 190(8): 3854-8, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23467935

RESUMO

Memory CD8 T cells recognizing conserved proteins from influenza A virus (IAV), such as nucleoprotein, have the potential to provide protection in individuals who lack the proper neutralizing Abs. In this study, we show that the most potent CD8 T cell-inducing influenza vaccine on the market (Flumist) does not induce sufficient numbers of cross-reactive CD8 T cells to provide substantial protection against lethal nonhomologous IAV challenge. However, Flumist-primed CD8 T cells rapidly acquire memory characteristics and can respond to short-interval boosting to greatly enlarge the IAV-specific memory pool, which is sufficient to protect mice from nonhomologous IAV challenge. Thus, a current vaccine strategy, Flumist, may serve as a priming platform for the rapid induction of large numbers of memory CD8 T cells with the capacity for broad protection against influenza.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Memória Imunológica , Vacinas contra Influenza/administração & dosagem , Influenza Humana/prevenção & controle , Ativação Linfocitária/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Reações Cruzadas/imunologia , Feminino , Humanos , Vírus da Influenza A/imunologia , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/uso terapêutico , Influenza Humana/imunologia , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos BALB C , Fatores de Tempo
12.
Proc Natl Acad Sci U S A ; 108(30): 12419-24, 2011 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-21746921

RESUMO

Inflammasomes are intracellular multiprotein signaling complexes that activate Caspase-1, leading to the cleavage and secretion of IL-1ß and IL-18, and ultimately host cell death. Inflammasome activation is a common cellular response to infection; however, the consequences of inflammasome activation during acute infection and in the development of long-term protective immunity is not well understood. To investigate the role of the inflammasome in vivo, we engineered a strain of Listeria monocytogenes that ectopically expresses Legionella pneumophila flagellin, a potent activator of the Nlrc4 inflammasome. Compared with wild-type L. monocytogenes, strains that ectopically secreted flagellin induced robust host cell death and IL-1ß secretion. These strains were highly attenuated both in bone marrow-derived macrophages and in vivo compared with wild-type L. monocytogenes. Attenuation in vivo was dependent on Nlrc4, but independent of IL-1ß/IL-18 or neutrophil activity. L. monocytogenes strains that activated the inflammasome generated significantly less protective immunity, a phenotype that correlated with decreased induction of antigen-specific T cells. Our data suggest that avoidance of inflammasome activation is a critical virulence strategy for intracellular pathogens, and that activation of the inflammasome leads to decreased long-term protective immunity and diminished T-cell responses.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/imunologia , Inflamassomos/genética , Inflamassomos/imunologia , Listeria monocytogenes/genética , Listeria monocytogenes/imunologia , Animais , Apoptose/imunologia , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Adaptadoras de Sinalização CARD , Proteínas de Ligação ao Cálcio/deficiência , Proteínas do Citoesqueleto/imunologia , Feminino , Flagelina/genética , Flagelina/imunologia , Engenharia Genética , Interações Hospedeiro-Patógeno/imunologia , Imunidade Celular , Imunidade Inata , Imunização , Legionella pneumophila/genética , Legionella pneumophila/imunologia , Listeria monocytogenes/patogenicidade , Listeriose/imunologia , Listeriose/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Linfócitos T/imunologia , Virulência/genética , Virulência/imunologia
13.
PLoS Pathog ; 7(3): e1002005, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21408203

RESUMO

Listeria monocytogenes is a facultative intracellular bacterial pathogen that can infect the placenta, a chimeric organ made of maternal and fetal cells. Extravillous trophoblasts (EVT) are specialized fetal cells that invade the uterine implantation site, where they come into direct contact with maternal cells. We have shown previously that EVT are the preferred site of initial placental infection. In this report, we infected primary human EVT with L. monocytogenes. EVT eliminated ∼80% of intracellular bacteria over 24-hours. Bacteria were unable to escape into the cytoplasm and remained confined to vacuolar compartments that became acidified and co-localized with LAMP1, consistent with bacterial degradation in lysosomes. In human placental organ cultures bacterial vacuolar escape rates differed between specific trophoblast subpopulations. The most invasive EVT-those that would be in direct contact with maternal cells in vivo-had lower escape rates than trophoblasts that were surrounded by fetal cells and tissues. Our results suggest that EVT present a bottleneck in the spread of L. monocytogenes from mother to fetus by inhibiting vacuolar escape, and thus intracellular bacterial growth. However, if L. monocytogenes is able to spread beyond EVT it can find a more hospitable environment. Our results elucidate a novel aspect of the maternal-fetal barrier.


Assuntos
Listeria monocytogenes/crescimento & desenvolvimento , Listeria monocytogenes/patogenicidade , Placenta/citologia , Placenta/microbiologia , Trofoblastos/citologia , Trofoblastos/microbiologia , Células Cultivadas , Feminino , Feto/microbiologia , Imunofluorescência , Humanos , Transmissão Vertical de Doenças Infecciosas , Lisossomos/metabolismo , Microscopia Eletrônica de Transmissão , Gravidez , Técnicas de Cultura de Tecidos , Útero/microbiologia , Vacúolos/microbiologia
14.
PLoS Pathog ; 7(3): e1001326, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21455492

RESUMO

Host defense against the intracellular pathogen Listeria monocytogenes (Lm) requires innate and adaptive immunity. Here, we directly imaged immune cell dynamics at Lm foci established by dendritic cells in the subcapsular red pulp (scDC) using intravital microscopy. Blood borne Lm rapidly associated with scDC. Myelomonocytic cells (MMC) swarmed around non-motile scDC forming foci from which blood flow was excluded. The depletion of scDC after foci were established resulted in a 10-fold reduction in viable Lm, while graded depletion of MMC resulted in 30-1000 fold increase in viable Lm in foci with enhanced blood flow. Effector CD8+ T cells at sites of infection displayed a two-tiered reduction in motility with antigen independent and antigen dependent components, including stable interactions with infected and non-infected scDC. Thus, swarming MMC contribute to control of Lm prior to development of T cell immunity by direct killing and sequestration from blood flow, while scDC appear to promote Lm survival while preferentially interacting with CD8+ T cells in effector sites.


Assuntos
Imunidade Adaptativa , Imunidade Inata , Listeria monocytogenes/patogenicidade , Listeriose/patologia , Linfócitos T Citotóxicos/patologia , Animais , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Células Dendríticas/patologia , Feminino , Técnicas de Introdução de Genes , Interações Hospedeiro-Patógeno , Listeriose/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Monócitos/patologia , Linfócitos T Citotóxicos/imunologia
15.
J Immunol ; 187(6): 2923-31, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21832161

RESUMO

One goal of immunization is to generate memory CD8 T cells of sufficient quality and quantity to confer protection against infection. It has been shown that memory CD8 T cell differentiation in vivo is controlled, at least in part, by the amount and duration of infection, Ag, and inflammatory cytokines present early after the initiation of the response. In this study, we used models of anti-vectorial immunity to investigate the impact of pre-existing immunity on the development and differentiation of vector-induced primary CD8 T cell responses. We showed that existing CD8 T cell memory influences the magnitude of naive CD8 T cell responses. However, the differentiation of newly recruited (either TCR-transgenic or endogenous) primary CD8 T cells into populations with the phenotype (CD62L(hi), CD27(hi), KLRG-1(low)) and function (tissue distribution, Ag-driven proliferation, cytokine production) of long-term memory was facilitated when they were primed in the presence of vector-specific memory CD8 T cells of the same or unrelated specificity. Therefore, these data suggested that the presence of anti-vectorial immunity impacts the rate of differentiation of vector-induced naive CD8 T cells, a notion with important implications for the design of future vaccination strategies.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Memória Imunológica/imunologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/citologia , Citocinas/biossíntese , Citocinas/imunologia , Imunofenotipagem , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo
16.
Ann Surg Oncol ; 19 Suppl 3: S597-607, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21979110

RESUMO

PURPOSE: Despite advances in therapy for the treatment of metastatic colorectal cancer, many patients die of hepatic disease. Current immunotherapeutic strategies are likely limited by inhibitory signals from the tumor. To successfully eliminate tumor deposits within an organ, an appropriate immunologic milieu to amplify antitumor responses must be developed. METHODS: We used a murine model utilizing the CT26 colon cancer cell line to analyze primary and memory tumor-specific T-cell responses induced by an attenuated actin A and internalin B deleted immunodominant tumor-associated antigen expressing strain of Listeria monocytogenes for the treatment of metastatic colorectal cancer. RESULTS: Treatment of mice bearing established hepatic metastases with this L. monocytogenes strain led to the generation of a strong initial tumor-specific cytotoxic CD8(+) T-cell response that successfully treated 90% of animals. Tumor antigen-specific central and effector memory T cells were also generated and protected against tumor rechallenge. These cell populations, when measured before and after tumor rechallenge, showed a marked expansion of antigen-specific effector CD8(+) effector memory T cells. This strain of L. monocytogenes was able to down-modulate the expression of the immune checkpoint molecule, PD-1, within the tumor microenvironment but had variable effects on CTLA-4 expression. CONCLUSIONS: This L. monocytogenes strain generated a highly effective antitumor T-cell response, providing a basis for the development of this vaccine platform in patients with liver metastases.


Assuntos
Antígenos de Neoplasias , Linfócitos T CD8-Positivos/metabolismo , Carcinoma/terapia , Neoplasias do Colo/terapia , Imunoterapia , Listeria monocytogenes/imunologia , Neoplasias Hepáticas/terapia , Análise de Variância , Animais , Antígenos de Diferenciação/metabolismo , Antineoplásicos Alquilantes/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Antígeno CTLA-4/metabolismo , Carcinoma/imunologia , Carcinoma/secundário , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Ciclofosfamida/uso terapêutico , Feminino , Interferon gama/metabolismo , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/secundário , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Receptor de Morte Celular Programada 1 , Estatísticas não Paramétricas , Análise de Sobrevida , Linfócitos T Reguladores
17.
Prostate Cancer Prostatic Dis ; 25(2): 219-228, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34257408

RESUMO

BACKGROUND: The safety and immunogenicity of JNJ-64041809 (JNJ-809), a live-attenuated, double-deleted Listeria monocytogenes (LADD Lm)-based immunotherapy targeting 4 relevant prostate cancer antigens, was evaluated in a phase 1 study in patients with metastatic castration-resistant prostate cancer (mCRPC). METHODS: Men with progressive mCRPC who had received ≥2 prior approved therapies were enrolled. Primary study objectives were to determine the recommended phase 2 dose (RP2D) and to evaluate the safety and immunogenicity of JNJ-809. RESULTS: A total of 26 patients received JNJ-809 (1 × 108 CFU (n = 6); 1 × 109 CFU (n = 20)). No dose-limiting toxicities were reported, and 1 × 109 CFU was selected as the RP2D. The most common adverse events (AEs) reported were chills (92%), pyrexia (81%), and fatigue (62%). The most frequent grade ≥3 AEs were lymphopenia (27%) and hypertension (23%). Serious AEs were reported in 27% of patients including 1 patient with grade 3 intestinal obstruction. JNJ-809 transiently induced peripheral cytokines, including interferon-γ, interleukin-10, and tumor necrosis factor-α. Of the 7 patients evaluable for T cell responses at the 1 × 109 CFU dose, evidence of post-treatment antigenic responses were observed in 6 to the Listeria antigen listeriolysin O and in 5 to ≥1 of the 4 encoded tumor antigens. Best overall response was stable disease in 13/25 response-evaluable patients. The study was terminated early as data collected were considered sufficient to evaluate safety and immunogenicity. CONCLUSIONS: JNJ-809 has manageable safety consistent with other LADD Lm-based therapies. Limited antigen-specific immune responses were observed, which did not translate into objective clinical responses.


Assuntos
Listeria monocytogenes , Neoplasias de Próstata Resistentes à Castração , Humanos , Imunoterapia/efeitos adversos , Listeria monocytogenes/genética , Masculino , Neoplasias de Próstata Resistentes à Castração/patologia
18.
J Clin Invest ; 118(12): 3990-4001, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19033668

RESUMO

Vaccine strategies that utilize human DCs to enhance antitumor immunity have yet to realize their full potential. Approaches that optimally target a spectrum of antigens to DCs are urgently needed. Here we report the development of a platform for loading DCs with antigen. It is based on killed but metabolically active (KBMA) recombinant Listeria monocytogenes and facilitates both antigen delivery and maturation of human DCs. Highly attenuated KBMA L. monocytogenes were engineered to express an epitope of the melanoma-associated antigen MelanA/Mart-1 that is recognized by human CD8+ T cells when presented by the MHC class I molecule HLA-A*0201. The engineered KBMA L. monocytogenes induced human DC upregulation of costimulatory molecules and secretion of pro-Th1 cytokines and type I interferons, leading to effective priming of Mart-1-specific human CD8+ T cells and lysis of patient-derived melanoma cells. KBMA L. monocytogenes expressing full-length NY-ESO-1 protein, another melanoma-associated antigen, delivered the antigen for presentation by MHC class I and class II molecules independent of the MHC haplotype of the DC donor. A mouse therapeutic tumor model was used to show that KBMA L. monocytogenes efficiently targeted APCs in vivo to induce protective antitumor responses. Together, our data demonstrate that KBMA L. monocytogenes may be a powerful platform that can both deliver recombinant antigen to DCs for presentation and provide a potent DC-maturation stimulus, making it a potential cancer vaccine candidate.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Listeria monocytogenes/imunologia , Melanoma/imunologia , Proteínas de Neoplasias/imunologia , Animais , Apresentação de Antígeno/genética , Antígenos de Neoplasias/genética , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/imunologia , Antígenos HLA-A/genética , Antígenos HLA-A/imunologia , Antígeno HLA-A2 , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Listeria monocytogenes/genética , Antígeno MART-1 , Melanoma/genética , Melanoma/terapia , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Neoplasias/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Células Th1/imunologia
19.
Environ Sci Technol ; 45(24): 10394-400, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22044020

RESUMO

The modification of emissions of climate-sensitive exhaust compounds such as CO(2), NO(x), hydrocarbons, and particulate matter from medium-speed marine diesel engines was studied for a set of fossil and biogenic fuels. Applied fossil fuels were the reference heavy fuel oil (HFO) and the low-sulfur marine gas oil (MGO); biogenic fuels were palm oil, soybean oil, sunflower oil, and animal fat. Greenhouse gas (GHG) emissions related to the production of biogenic fuels were treated by means of a fuel life cycle analysis which included land use changes associated with the growth of energy plants. Emissions of CO(2) and NO(x) per kWh were found to be similar for fossil fuels and biogenic fuels. PM mass emission was reduced to 10-15% of HFO emissions for all low-sulfur fuels including MGO as a fossil fuel. Black carbon emissions were reduced significantly to 13-30% of HFO. Changes in emissions were predominantly related to particulate sulfate, while differences between low-sulfur fossil fuels and low-sulfur biogenic fuels were of minor significance. GHG emissions from the biogenic fuel life cycle (FLC) depend crucially on energy plant production conditions and have the potential of shifting the overall GHG budget from positive to negative compared to fossil fuels.


Assuntos
Poluentes Atmosféricos/análise , Biocombustíveis , Navios/métodos , Emissões de Veículos/análise , Poluição do Ar/estatística & dados numéricos , Efeito Estufa , Navios/instrumentação
20.
Proc Natl Acad Sci U S A ; 105(29): 10191-6, 2008 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-18632558

RESUMO

To gain insight into the interaction of intracellular pathogens with host innate immune pathways, we performed an unbiased genetic screen of Listeria monocytogenes mutants that induced an enhanced or diminished host innate immune response. Here, we show that the major facilitator superfamily of bacterial multidrug resistance transporters (MDRs) controlled the magnitude of a host cytosolic surveillance pathway, leading to the production of several cytokines, including type I IFN. Mutations mapping to repressors of MDRs resulted in ectopic expression of their cognate transporters, leading to host responses that were increased up to 20-fold over wild-type bacteria, and a 20-fold decrease in bacterial growth in vivo. Mutation of one of the MDRs, MdrM, led to a 3-fold reduction in the IFN-beta response to L. monocytogenes infection, indicating a pivotal role for MdrM in activation of the host cytosolic surveillance system. Bacterial MDRs had previously been associated with resistance to antibiotics and other toxic compounds. This report links bacterial MDRs and host immunity. Understanding the mechanisms through which live pathogens activate innate immune signaling pathways should lead to the discovery of adjuvants, vaccines, and perhaps new classes of therapeutics. Indeed, we show that the mutants identified in this screen induced vastly altered type I IFN response in vivo as well.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/imunologia , Proteínas de Bactérias/imunologia , Imunidade Inata , Listeria monocytogenes/imunologia , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Animais , Proteínas de Bactérias/genética , Citosol/imunologia , Genes Bacterianos , Genes MDR , Interferon beta/biossíntese , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidade , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA