Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Res ; 84(7): 1029-1047, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38270915

RESUMO

The clinical benefits of tumor-targeting antibodies (tAb) are modest in solid human tumors. The efficacy of many tAbs is dependent on Fc receptor (FcR)-expressing leukocytes that bind Fc fragments of tAb. Tumor-associated macrophages (TAM) and neutrophils (TAN) represent the majority of FcR+ effectors in solid tumors. A better understanding of the mechanisms by which TAMs and TANs regulate tAb response could help improve the efficacy of cancer treatments. Here, we found that myeloid effectors interacting with tAb-opsonized lung cancer cells used antibody-dependent trogocytosis (ADT) but not antibody-dependent phagocytosis. During this process, myeloid cells "nibbled off" tumor cell fragments containing tAb/targeted antigen (tAg) complexes. ADT was only tumoricidal when the tumor cells expressed high levels of tAg and the effectors were present at high effector-to-tumor ratios. If either of these conditions were not met, which is typical for solid tumors, ADT was sublethal. Sublethal ADT, mainly mediated by CD32hiCD64hi TAM, led to two outcomes: (i) removal of surface tAg/tAb complexes from the tumor that facilitated tumor cell escape from the tumoricidal effects of tAb; and (ii) acquisition of bystander tAgs by TAM with subsequent cross-presentation and stimulation of tumor-specific T-cell responses. CD89hiCD32loCD64lo peripheral blood neutrophils (PBN) and TAN stimulated tumor cell growth in the presence of the IgG1 anti-EGFR Ab cetuximab; however, IgA anti-EGFR Abs triggered the tumoricidal activity of PBN and negated the stimulatory effect of TAN. Overall, this study provides insights into the mechanisms by which myeloid effectors mediate tumor cell killing or resistance during tAb therapy. SIGNIFICANCE: The elucidation of the conditions and mechanisms by which human FcR+ myeloid effectors mediate cancer cell resistance and killing during antibody treatment could help develop improved strategies for treating solid tumors.


Assuntos
Neoplasias , Neutrófilos , Humanos , Neutrófilos/metabolismo , Macrófagos Associados a Tumor/metabolismo , Trogocitose , Citotoxicidade Celular Dependente de Anticorpos , Fagocitose , Neoplasias/patologia , Receptores Fc , Antígenos de Neoplasias
2.
Mol Ther ; 29(2): 658-670, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33160076

RESUMO

Gene-mediated cytotoxic immunotherapy (GMCI) is an immuno-oncology approach involving local delivery of a replication-deficient adenovirus expressing herpes simplex thymidine kinase (AdV-tk) followed by anti-herpetic prodrug activation that promotes immunogenic tumor cell death, antigen-presenting cell activation, and T cell stimulation. This phase I dose-escalation pilot trial assessed bronchoscopic delivery of AdV-tk in patients with suspected lung cancer who were candidates for surgery. A single intra-tumoral AdV-tk injection in three dose cohorts (maximum 1012 viral particles) was performed during diagnostic staging, followed by a 14-day course of the prodrug valacyclovir, and subsequent surgery 1 week later. Twelve patients participated after appropriate informed consent. Vector-related adverse events were minimal. Immune biomarkers were evaluated in tumor and blood before and after GMCI. Significantly increased infiltration of CD8+ T cells was found in resected tumors. Expression of activation, inhibitory, and proliferation markers, such as human leukocyte antigen (HLA)-DR, CD38, Ki67, PD-1, CD39, and CTLA-4, were significantly increased in both the tumor and peripheral CD8+ T cells. Thus, intratumoral AdV-tk injection into non-small-cell lung cancer (NSCLC) proved safe and feasible, and it effectively induced CD8+ T cell activation. These data provide a foundation for additional clinical trials of GMCI for lung cancer patients with potential benefit if combined with other immune therapies.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Terapia Genética , Imunoterapia/métodos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/terapia , Adenoviridae/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Citotoxicidade Imunológica , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Neoplasias Pulmonares/patologia , Terapia Neoadjuvante , Timidina Quinase/genética
3.
Cell Rep ; 33(11): 108500, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33326785

RESUMO

Immune cell function is influenced by metabolic conditions. Low-glucose, high-lactate environments, such as the placenta, gastrointestinal tract, and the tumor microenvironment, are immunosuppressive, especially for glycolysis-dependent effector T cells. We report that nicotinamide adenine dinucleotide (NAD+), which is reduced to NADH by lactate dehydrogenase in lactate-rich conditions, is a key point of metabolic control in T cells. Reduced NADH is not available for NAD+-dependent enzymatic reactions involving glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and 3-phosphoglycerate dehydrogenase (PGDH). We show that increased lactate leads to a block at GAPDH and PGDH, leading to the depletion of post-GAPDH glycolytic intermediates, as well as the 3-phosphoglycerate derivative serine that is known to be important for T cell proliferation. Supplementing serine rescues the ability of T cells to proliferate in the presence of lactate-induced reductive stress. Directly targeting the redox state may be a useful approach for developing novel immunotherapies in cancer and therapeutic immunosuppression.


Assuntos
Ácido Láctico/metabolismo , NAD/metabolismo , Proliferação de Células , Humanos , Oxirredução
4.
Mol Imaging Biol ; 22(1): 144-155, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31820349

RESUMO

PURPOSE: Intraoperative molecular imaging (IMI) utilizes optical dyes that accumulate within tumors to assist with detection during a cancer operation. IMI can detect disease not visualized preoperatively, as well as positive margins. However, these dyes are limited by autofluorescence, signal reflection, and photon-scatter. We hypothesize that a novel dye with a wide separation between excitation and emission spectra, SS180, would help overcome these obstacles. PROCEDURES: Two targeted molecular contrast agents, OTL38 and SS180, were selected for this study. Both dyes had the same targeting ligand to folate receptor alpha (FRα). OTL38, a well-annotated IMI agent in human trials, has a Stokes shift of 22 nm, whereas SS180, the new dye, has a Stokes shift of 129 nm. Cell lines were tested for FRα expression and incubated with dyes to demonstrate receptor-dependent binding. Cells were incubated in various concentrations of the dyes to compare dose- and time-dependent binding. Finally, cells tagged with the dyes were injected subcutaneously in a murine model to estimate tumor burden necessary to generate fluorescent signal. RESULTS: Cellular studies demonstrated that SS180 binds cells in a dose-, receptor-, and time-dependent manner and exhibits higher mean fluorescence intensities by flow cytometry when compared with OTL38 for each time point and concentration. In an in vivo flank tumor model, SS180 had a higher tumor-to-background ratio (TBR) than OTL38, though not statistically significant (p = 0.08). Ex vivo, OTL38 had a higher TBR than SS180 (p = 0.02). The subcutaneous model revealed that SS180 had a higher TBR at 5 × 106 cells than OTL38 (p = 0.05). No toxicity was observed in the animals. CONCLUSIONS: SS180 exhibits greater TBRs in vivo, but not ex vivo. These findings suggest that SS180 may have weaker fluorescence, but superior contrast. Studies in large animal models and clinical trials may better elucidate the clinical value of a long Stokes shift.


Assuntos
Fluorescência , Corantes Fluorescentes/farmacocinética , Receptor 1 de Folato/metabolismo , Imagem Molecular/métodos , Neoplasias/cirurgia , Cirurgia Assistida por Computador/métodos , Animais , Linhagem Celular Tumoral , Corantes Fluorescentes/química , Humanos , Cuidados Intraoperatórios , Camundongos , Camundongos Nus , Neoplasias/diagnóstico por imagem , Neoplasias/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Oncoimmunology ; 8(9): e1638211, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31428531

RESUMO

Given the growing interest and promising preliminary results of immunotherapy in malignant pleural mesothelioma (MPM), it has become important to more fully understand the immune landscape in this tumor. This may be especially relevant in deciding who might benefit most from checkpoint blockade or agonist antibody therapy. Since the phenotype of tumor infiltrating lymphocytes (TILs) in MPM has not been fully described and their function has not been carefully assessed, we collected fresh tumor and blood from 22 patients undergoing surgical resection and analysed single cell suspensions by flow cytometry. The functionality of TILs was assessed by measurement of cytokine expression (IFN-γ) following overnight stimulation ex vivo. Results showed low numbers of CD8+ TILs whose function was either moderately or severely suppressed. The degree of TIL hypofunction did not correlate with the presence of co-existing macrophages or neutrophils, nor with expression of the inhibitory receptors PD-1, CD39 and CTLA-4. Hypofunction was associated with higher numbers of CD4 regulatory T cells (Tregs) and with expression of the inhibitory receptor TIGIT. On the other hand, presence of tissue-resident memory (Trm) cells and expression of TIM-3 on CD8+ cells were positively associated with cytokine production. However, Trm function was partially suppressed when the transcription factor Eomesodermin (Eomes) was co-expressed. Understanding the function of TILs in malignant mesothelioma may have clinical implications for immunotherapy, especially in choosing the best immunotherapy targets. Our data suggests that Treg cell blocking agents or TIGIT inhibitor antibodies might be especially valuable in these patients.

6.
Sci Transl Med ; 11(479)2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30760579

RESUMO

Data from mouse tumor models suggest that tumor-associated monocyte/macrophage lineage cells (MMLCs) dampen antitumor immune responses. However, given the fundamental differences between mice and humans in tumor evolution, genetic heterogeneity, and immunity, the function of MMLCs might be different in human tumors, especially during early stages of disease. Here, we studied MMLCs in early-stage human lung tumors and found that they consist of a mixture of classical tissue monocytes and tumor-associated macrophages (TAMs). The TAMs coexpressed M1/M2 markers, as well as T cell coinhibitory and costimulatory receptors. Functionally, TAMs did not primarily suppress tumor-specific effector T cell responses, whereas tumor monocytes tended to be more T cell inhibitory. TAMs expressing relevant MHC class I/tumor peptide complexes were able to activate cognate effector T cells. Mechanistically, programmed death-ligand 1 (PD-L1) expressed on bystander TAMs, as opposed to PD-L1 expressed on tumor cells, did not inhibit interactions between tumor-specific T cells and tumor targets. TAM-derived PD-L1 exerted a regulatory role only during the interaction of TAMs presenting relevant peptides with cognate effector T cells and thus may limit excessive activation of T cells and protect TAMs from killing by these T cells. These results suggest that the function of TAMs as primarily immunosuppressive cells might not fully apply to early-stage human lung cancer and might explain why some patients with strong PD-L1 positivity fail to respond to PD-L1 therapy.


Assuntos
Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Macrófagos/patologia , Monócitos/patologia , Linfócitos T/imunologia , Células A549 , Antígenos de Neoplasias/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Comunicação Celular , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Receptores de Lipopolissacarídeos/metabolismo , Proteínas de Neoplasias/metabolismo , Estadiamento de Neoplasias , Peptídeos/metabolismo , Fenótipo , Transdução de Sinais
7.
Mol Ther ; 26(2): 390-403, 2018 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-29241970

RESUMO

Non-small cell lung cancer (NSCLC) is the number one cancer killer in the United States. Despite attempted curative surgical resection, nearly 40% of patients succumb to recurrent disease. High recurrence rates may be partially explained by data suggesting that 20% of NSCLC patients harbor synchronous disease that is missed during resection. In this report, we describe the use of a novel folate receptor-targeted near-infrared contrast agent (OTL38) to improve the intraoperative localization of NSCLC during pulmonary resection. Using optical phantoms, fluorescent imaging with OTL38 was associated with less autofluorescence and greater depth of detection compared to traditional optical contrast agents. Next, in in vitro and in vivo NSCLC models, OTL38 reliably localized NSCLC models in a folate receptor-dependent manner. Before testing intraoperative molecular imaging with OTL38 in humans, folate receptor-alpha expression was confirmed to be present in 86% of pulmonary adenocarcinomas upon histopathologic review of 100 human pulmonary resection specimens. Lastly, in a human feasibility study, intraoperative molecular imaging with OTL38 accurately identified 100% of pulmonary adenocarcinomas and allowed for identification of additional subcentimeter neoplastic processes in 30% of subjects. This technology may enhance the surgeon's ability to identify NSCLC during oncologic resection and potentially improve long-term outcomes.


Assuntos
Adenocarcinoma/diagnóstico por imagem , Adenocarcinoma/metabolismo , Meios de Contraste , Receptores de Folato com Âncoras de GPI/metabolismo , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/metabolismo , Imagem Molecular , Adenocarcinoma/genética , Adenocarcinoma/cirurgia , Animais , Modelos Animais de Doenças , Feminino , Receptor 1 de Folato/genética , Receptor 1 de Folato/metabolismo , Receptores de Folato com Âncoras de GPI/genética , Expressão Gênica , Humanos , Imuno-Histoquímica , Cuidados Intraoperatórios , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/cirurgia , Masculino , Camundongos , Imagem Molecular/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
JCI Insight ; 2(16)2017 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-28814673

RESUMO

Experimental data indicate that FOXP3+ Tregs can markedly curtail host antitumor immune responses, but the properties of human intratumoral Tregs are still largely unknown, in part due to significant methodologic problems. We studied the phenotypic, functional, epigenetic, and transcriptional features of Tregs in 92 patients with non-small-cell lung cancer, comparing the features of Tregs within tumors versus corresponding blood, lung, and lymph node samples. Intratumoral Treg numbers and suppressive function were significantly increased compared with all other sites but did not display a distinctive phenotype by flow cytometry. However, by undertaking simultaneous evaluation of mRNA and protein expression at the single-cell level, we demonstrated that tumor Tregs have a phenotype characterized by upregulated expression of FOXP3 mRNA and protein as well as significantly increased expression of EOS, IRF4, SATB1, and GATA1 transcription factor mRNAs. Expression of these "Treg-locking" transcription factors was positively correlated with levels of FOXP3 mRNA, with highest correlations for EOS and SATB1. EOS had an additional, FOXP3 mRNA-independent, positive correlation with FOXP3 protein in tumor Tregs. Our study identifies distinctive features of intratumoral Tregs and suggests that targeting Treg-locking transcription factors, especially EOS, may be of clinical importance for antitumor Treg-based therapy.

9.
J Immunol ; 197(6): 2280-9, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27489283

RESUMO

Although ribosomal proteins (RP) are thought to primarily facilitate biogenesis of the ribosome and its ability to synthesize protein, emerging evidence suggests that individual RP can perform critical regulatory functions that control developmental processes. We showed previously that despite the ubiquitous expression of the RP ribosomal protein L22 (Rpl22), germline ablation of Rpl22 in mice causes a selective, p53-dependent block in the development of αß, but not γδ, T cell progenitors. Nevertheless, the basis by which Rpl22 loss selectively induces p53 in αß T cell progenitors remained unclear. We show in this study that Rpl22 regulates the development of αß T cells by restraining endoplasmic reticulum (ER) stress responses. In the absence of Rpl22, ER stress is exacerbated in αß, but not γδ, T cell progenitors. The exacerbated ER stress in Rpl22-deficient αß T lineage progenitors is responsible for selective induction of p53 and their arrest, as pharmacological induction of stress is sufficient to induce p53 and replicate the selective block of αß T cells, and attenuation of ER stress signaling by knockdown of protein kinase R-like ER kinase, an ER stress sensor, blunts p53 induction and rescues development of Rpl22-deficient αß T cell progenitors. Rpl22 deficiency appears to exacerbate ER stress by interfering with the ability of ER stress signals to block new protein synthesis. Our finding that Rpl22 deficiency exacerbates ER stress responses and induces p53 in αß T cell progenitors provides insight into how a ubiquitously expressed RP can perform regulatory functions that are selectively required by some cell lineages but not others.


Assuntos
Estresse do Retículo Endoplasmático , Regulação da Expressão Gênica , Células Precursoras de Linfócitos T/fisiologia , Proteínas de Ligação a RNA/fisiologia , Receptores de Antígenos de Linfócitos T alfa-beta , Proteínas Ribossômicas/fisiologia , Transdução de Sinais , Subpopulações de Linfócitos T/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula/fisiologia , Camundongos , Proteínas Ribossômicas/deficiência , Subpopulações de Linfócitos T/imunologia , Proteína Supressora de Tumor p53/metabolismo
10.
Cancer Res ; 76(11): 3387-96, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27197189

RESUMO

Mutations in ribosomal proteins cause bone marrow failure syndromes associated with increased cancer risk, but the basis by which they do so remains unclear. We reported previously that the ribosomal protein Rpl22 is a tumor suppressor in T-cell acute lymphoblastic leukemia/lymphoma (T-ALL), and that loss of just one Rpl22 allele accelerates T-cell lymphomagenesis by activating NF-κB and inducing the stem cell factor Lin28B. Here, we show that, paradoxically, loss of both alleles of Rpl22 restricts lymphoma progression through a distinct effect on migration of malignant cells out of the thymus. Lymphoma-prone AKT2-transgenic or PTEN-deficient mice on an Rpl22(-/-) background developed significantly larger and markedly more vascularized thymic tumors than those observed in Rpl22(+/+) control mice. But, unlike Rpl22(+/+) or Rpl22(+/-) tumors, Rpl22(-/-) lymphomas did not disseminate to the periphery and were retained in the thymus. We traced the defect in the Rpl22(-/-) lymphoma migratory capacity to downregulation of the KLF2 transcription factor and its targets, including the key migratory factor sphingosine 1-phosphate receptor 1 (S1PR1). Indeed, reexpression of S1PR1 in Rpl22-deficient tumor cells restores their migratory capacity in vitro The regulation of KLF2 and S1PR1 by Rpl22 appears to be proximal as Rpl22 reexpression in Rpl22-deficient lymphoma cells restores expression of KLF2 and S1P1R, while Rpl22 knockdown in Rpl22-sufficient lymphomas attenuates their expression. Collectively, these data reveal that, while loss of one copy of Rpl22 promotes lymphomagenesis and disseminated disease, loss of both copies impairs responsiveness to migratory cues and restricts malignant cells to the thymus. Cancer Res; 76(11); 3387-96. ©2016 AACR.


Assuntos
Linfoma de Células T/patologia , PTEN Fosfo-Hidrolase/fisiologia , Proteínas de Ligação a RNA/fisiologia , Proteínas Ribossômicas/fisiologia , Linfócitos T/patologia , Neoplasias do Timo/secundário , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Movimento Celular , Proliferação de Células , Feminino , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Linfoma de Células T/genética , Linfoma de Células T/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/genética , NF-kappa B/metabolismo , Invasividade Neoplásica , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/metabolismo , Neoplasias do Timo/genética , Neoplasias do Timo/metabolismo , Células Tumorais Cultivadas
11.
J Immunol ; 187(2): 664-75, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21690328

RESUMO

αß and γδ lineage T cells are thought to arise from a common CD4(-)CD8(-) progenitor in the thymus. However, the molecular pathways controlling fate selection and maturation of these two lineages remain poorly understood. We demonstrated recently that a ubiquitously expressed ribosomal protein, Rpl22, is selectively required for the development of αß lineage T cells. Germline ablation of Rpl22 impairs development of αß lineage, but not γδ lineage, T cells through activation of a p53-dependent checkpoint. In this study, we investigate the downstream effectors used by p53 to impair T cell development. We found that many p53 targets were induced in Rpl22(-/-) thymocytes, including miR-34a, PUMA, p21(waf), Bax, and Noxa. Notably, the proapoptotic factor Bim, while not a direct p53 target, was also strongly induced in Rpl22(-/-) T cells. Gain-of-function analysis indicated that overexpression of miR-34a caused a developmental arrest reminiscent of that induced by p53 in Rpl22-deficient T cells; however, only a few p53 targets alleviated developmental arrest when individually ablated by gene targeting or knockdown. Co-elimination of PUMA and Bim resulted in a nearly complete restoration of development of Rpl22(-/-) thymocytes, indicating that p53-mediated arrest is enforced principally through effects on cell survival. Surprisingly, co-elimination of the primary p53 regulators of cell cycle arrest (p21(waf)) and apoptosis (PUMA) actually abrogated the partial rescue caused by loss of PUMA alone, suggesting that the G1 checkpoint protein p21(waf) facilitates thymocyte development in some contexts.


Assuntos
Diferenciação Celular/imunologia , Marcação de Genes , Inibidores do Crescimento/imunologia , Proteínas Ribossômicas/deficiência , Subpopulações de Linfócitos T/imunologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Apoptose/genética , Apoptose/imunologia , Proteínas Reguladoras de Apoptose/biossíntese , Proteínas Reguladoras de Apoptose/deficiência , Proteína 11 Semelhante a Bcl-2 , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Marcação de Genes/métodos , Inibidores do Crescimento/genética , Proteínas de Membrana/biossíntese , Proteínas de Membrana/deficiência , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/deficiência , Proteínas Ribossômicas/biossíntese , Proteínas Ribossômicas/genética , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Timo/imunologia , Timo/metabolismo , Timo/patologia , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/deficiência , Proteínas Supressoras de Tumor/biossíntese , Proteínas Supressoras de Tumor/deficiência , Proteína X Associada a bcl-2/biossíntese
12.
Semin Immunol ; 22(4): 237-46, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20471282

RESUMO

While insights into the molecular processes that specify adoption of the alphabeta and gammadelta fates are beginning to emerge, the basis for control of specification remains highly controversial. This review highlights the current models attempting to explain T lineage commitment. Recent observations support the hypothesis that the T cell receptor (TCR) provides instructive cues through differences in TCR signaling intensity and/or longevity. Accordingly, we review evidence addressing the importance of differences in signal strength/longevity, how signals differing in intensity/longevity may be generated, and finally how such signals modulate the activity of downstream effectors to promote the opposing developmental fates.


Assuntos
Linhagem da Célula , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Transdução de Sinais , Linfócitos T/citologia , Linfócitos T/imunologia , Animais , Humanos , Modelos Imunológicos , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T/metabolismo
13.
Blood ; 113(7): 1464-73, 2009 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-18791164

RESUMO

These studies investigate how interactions between the BCR and FcgammaRIIB affect B lymphocyte stimulator (BLyS) recep-tor expression and signaling. Previous studies showed that BCR ligation up-regulates BLyS binding capacity in mature B cells, reflecting increased BLyS receptor levels. Here we show that FcgammaRIIB coaggregation dampens BCR-induced BLyS receptor up-regulation. This cross-regulation requires BCR and FcgammaRIIB coligation, and optimal action relies on the Src-homology-2 (SH2)-containing inositol 5 phosphase-1 (SHIP1). Subsequent to FcgammaRIIB/BCR coaggregation, the survival promoting actions of BLyS are attenuated, reflecting reduced BLyS receptor signaling capacity in terms of Pim 2 maintenance, noncanonical NF-kappaB activation, and Bcl-xL levels. These findings link the negative regulatory functions of FcgammaRIIB with BLyS-mediated B-cell survival.


Assuntos
Fator Ativador de Células B/metabolismo , Linfócitos B/fisiologia , Proteínas Proto-Oncogênicas c-bcr/metabolismo , Receptores de IgG/metabolismo , Transdução de Sinais/imunologia , Animais , Fator Ativador de Células B/genética , Linfócitos B/citologia , Sobrevivência Celular/imunologia , Células Cultivadas , Feminino , Imunoglobulina D/metabolismo , Imunoglobulina M/metabolismo , Inositol Polifosfato 5-Fosfatases , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Receptores de IgG/genética , Regulação para Cima/imunologia , Proteína bcl-X/genética
14.
Cell Biochem Biophys ; 53(1): 1-16, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19034695

RESUMO

The B Lymphocyte Stimulator (BLyS) family of ligands and receptors regulates humoral immunity by controlling B lymphocyte survival and differentiation. Herein, we review the ligands and receptors of this family, their biological functions, and the biochemical processes through which they operate. Pre-immune B lymphocytes rely on BLyS signaling for their survival, whereas antigen experienced B lymphocytes generally interact more avidly with a homologous cytokine, A Proliferation Inducing Ligand (APRIL). The molecular basis for signaling via the three BLyS family receptors reveals complex interplay with other B lymphocyte signaling systems, affording the integration of selective and homeostatic processes. As our understanding of this system advances, molecular targets for manipulating humoral immunity in both health and disease should be revealed.


Assuntos
Fator Ativador de Células B/metabolismo , Linfócitos B/metabolismo , Homeostase/imunologia , Animais , Formação de Anticorpos/fisiologia , Fator Ativador de Células B/imunologia , Linfócitos B/citologia , Linfócitos B/imunologia , Diferenciação Celular/imunologia , Humanos , Ligantes , Ativação Linfocitária/imunologia , Transdução de Sinais/imunologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/imunologia
15.
Nat Immunol ; 9(12): 1379-87, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18978795

RESUMO

The survival of transitional and mature B cells requires both the B cell antigen receptor (BCR) and BLyS receptor 3 (BR3), which suggests that these receptors send signals that are nonredundant or that engage in crosstalk with each other. Here we show that BCR signaling induced production of the nonclassical transcription factor NF-kappaB pathway substrate p100, which is required for transmission of BR3 signals and thus B cell survival. The capacity for sustained p100 production emerged during transitional B cell differentiation, the stage at which BCR signals begin to mediate survival rather than negative selection. Our findings identify a molecular mechanism for the reliance of primary B cells on continuous BR3 and BCR signaling, as well as for the gradual resistance to negative selection that is acquired during B cell maturation.


Assuntos
Fator Ativador de Células B/metabolismo , Linfócitos B/citologia , Diferenciação Celular/imunologia , NF-kappa B/metabolismo , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais/imunologia , Animais , Fator Ativador de Células B/imunologia , Linfócitos B/imunologia , Linhagem Celular , Sobrevivência Celular/imunologia , Citometria de Fluxo , Humanos , Immunoblotting , Camundongos , NF-kappa B/imunologia , Receptor Cross-Talk/imunologia , Receptores de Antígenos de Linfócitos B/imunologia
16.
Immunol Res ; 42(1-3): 75-83, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18668213

RESUMO

Lymphocyte homeostasis poses a multi-faceted biological puzzle, because steady pre-immune populations must be maintained at an acceptable steady state to yield effective protection, despite stringent selective events during their generation. In addition, activated, memory and both short- and long-term effectors must be governed by independent homeostatic mechanisms. Finally, advancing age is accompanied by substantial changes that impact the dynamics and behavior of these pools, leading to cumulative homeostatic perturbations and compensation. Our laboratory has focused on the over-arching role of BLyS family ligands and receptors in these processes. These studies have led to a conceptual framework within which distinct homeostatic niches are specified by BLyS receptor signatures, which define the BLyS family ligands that can afford survival. The cues for establishing these receptor signatures, as well as the downstream survival mechanisms involved, are integrated with cell extrinsic inputs via cross talk among downstream mediators. A refined understanding of these relationships should yield insight into the selection and maintenance of B cell subsets, as well as an appreciation of how homeostatic mechanisms may contribute to immunosenescence.


Assuntos
Subpopulações de Linfócitos B/citologia , Homeostase/fisiologia , Animais , Fator Ativador de Células B/fisiologia , Receptor do Fator Ativador de Células B/fisiologia , Antígeno de Maturação de Linfócitos B/fisiologia , Senescência Celular/fisiologia , Receptores de Antígenos de Linfócitos B/fisiologia , Proteína Transmembrana Ativadora e Interagente do CAML/fisiologia , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia
17.
Curr Opin Immunol ; 20(2): 158-61, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18456486

RESUMO

BAFF and its receptors play a crucial role in peripheral B cell selection and survival, by dictating the set point for mature primary B cell numbers and adjusting thresholds for specificity-based selection during transitional differentiation. The notion that selective stringency can be varied on the basis of homeostatic demands reveals a previously unappreciated degree of plasticity in B cell tolerance, and suggests a paradigm that unites peripheral negative and positive selection with the maintenance of mature B cell numbers. Moreover, it implies a developmentally regulated coupling of BCR and BAFF receptors at the transitional stages and beyond.


Assuntos
Fator Ativador de Células B/metabolismo , Receptor do Fator Ativador de Células B/metabolismo , Linfócitos B/imunologia , Tolerância Imunológica , Animais , Humanos , Camundongos , Receptores de Antígenos de Linfócitos B/metabolismo , Transdução de Sinais
18.
J Immunol ; 178(12): 7531-9, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17548587

RESUMO

Through their differential interactions with B lymphocyte stimulator (BLyS) and a proliferation-inducing ligand (APRIL), the three BLyS family receptors play central roles in B cell survival and differentiation. Recent evidence indicates BLyS receptor levels shift following BCR ligation, suggesting that activation cues can alter overall BLyS receptor profiles and thus ligand sensitivity. In this study, we show that TLR stimuli also alter BLyS receptor expression, but in contrast to BCR ligation, TLR9 and TLR4 signals, preferentially increase transmembrane activator calcium modulator and cyclophilin ligand interactor (TACI) expression. Although both of these TLRs act through MyD88-dependent mechanisms to increase TACI expression, they differ in terms of their downstream mediators and the B cell subset affected. Surprisingly, only TLR4 relies on c-Rel and p50 to augment TACI expression, whereas TLR9 does not. Furthermore, although all follicular and marginal zone B cells up-regulate TACI in response to TLR9 stimulation, only marginal zone B cells and a subset of follicular B cells respond to TLR4. Finally, we find that both BLyS and APRIL enhance viability among quiescent and BCR-stimulated B cells. However, although BLyS enhances viability among TLR stimulated B cells, APRIL does not, suggesting that TACI but not BLyS receptor 3 may share survival promoting pathways with TLRs.


Assuntos
Linfócitos B/imunologia , Receptores do Fator de Necrose Tumoral/metabolismo , Receptor 4 Toll-Like/agonistas , Receptor Toll-Like 9/agonistas , Animais , Receptor do Fator Ativador de Células B/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/metabolismo , Oligodesoxirribonucleotídeos/farmacologia , Proteínas Proto-Oncogênicas c-rel/metabolismo , Proteína Transmembrana Ativadora e Interagente do CAML/metabolismo , Membro 13 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Regulação para Cima
19.
Immunity ; 25(3): 395-6, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16979572

RESUMO

Two recent Immunity articles (Enzler et al., 2006; Sasaki et al., 2006) probe the roles of Nuclear Factor kappa-B (NF-kappaB) pathways in survival and differentiation mediated by B cell activation factor of the TNF family (BAFF).


Assuntos
Subpopulações de Linfócitos B/citologia , Subpopulações de Linfócitos B/metabolismo , Diferenciação Celular/imunologia , Transdução de Sinais/imunologia , Animais , Sobrevivência Celular/imunologia
20.
J Immunol ; 176(11): 6405-10, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16709796

RESUMO

The BLyS family of ligands and receptors governs B cell homeostasis by controlling survival, differentiation, and lifespan. This family consists of multiple receptors and ligands, allowing independent regulation of different B cell subsets by varying the combination and levels of receptors expressed. Multiple downstream signaling pathways are implicated in these activities, reflecting this receptor complexity as well as cross-talk with other B cell signaling systems. BLyS levels are associated with multiple forms of humoral autoimmunity and can modulate tolerogenic elimination at the transitional checkpoint. BLyS responsiveness thus balances peripheral selection against cell numbers, providing an elastic system that varies selective stringency based on homeostatic demands.


Assuntos
Linfócitos B/citologia , Linfócitos B/imunologia , Diferenciação Celular/imunologia , Proteínas de Membrana/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Fator Ativador de Células B , Linfócitos B/metabolismo , Sobrevivência Celular/imunologia , Humanos , Proteínas de Membrana/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...