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











Base de dados
Intervalo de ano de publicação
1.
Immunol Cell Biol ; 102(1): 8-11, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37982351

RESUMO

The advent and clinical success of immune checkpoint inhibitors Ipilimumab, Nivolumab and Pembrolizumab has had a seismic impact on our drug discovery focus and rationale. Novel extrinsic targets that enhance immune responses to cancer are actively being pursued, while tumor intrinsic targets that render cancer cells more sensitive to the immune system have joined traditional intrinsic targets (e.g. directly cytotoxic) in the drug discovery pipeline. The phosphatase PTPN2 (TC-PTP) and its paralog PTPN1 (PTP-1B) are negative regulators of several cytokine signaling pathways and T cell receptor (TCR) signaling. In a recent publication, Baumgartner et al. demonstrate the pre-clinical efficacy of a first-in-class dual PTPN1/N2 active site inhibitor (ABBV-CLS-484/AC484) in cancer models.


Assuntos
Anticorpos Monoclonais , Neoplasias , Humanos , Anticorpos Monoclonais/uso terapêutico , Nivolumabe/uso terapêutico , Ipilimumab/uso terapêutico , Neoplasias/tratamento farmacológico , Imunoterapia , Biologia
2.
Cancer Cell ; 33(6): 1033-1047.e5, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29894690

RESUMO

The co-engagement of fragment crystallizable (Fc) gamma receptors (FcγRs) with the Fc region of recombinant immunoglobulin monoclonal antibodies (mAbs) and its contribution to therapeutic activity has been extensively studied. For example, Fc-FcγR interactions have been shown to be important for mAb-directed effector cell activities, as well as mAb-dependent forward signaling into target cells via receptor clustering. Here we identify a function of mAbs targeting T cell-expressed antigens that involves FcγR co-engagement on antigen-presenting cells (APCs). In the case of mAbs targeting CTLA-4 and TIGIT, the interaction with FcγR on APCs enhanced antigen-specific T cell responses and tumoricidal activity. This mechanism extended to an anti-CD45RB mAb, which led to FcγR-dependent regulatory T cell expansion in mice.


Assuntos
Anticorpos Monoclonais/imunologia , Células Apresentadoras de Antígenos/imunologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Receptores de IgG/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/uso terapêutico , Células Apresentadoras de Antígenos/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antígeno CTLA-4/imunologia , Antígeno CTLA-4/metabolismo , Humanos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/metabolismo , Ligação Proteica , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de IgG/metabolismo , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo
3.
PLoS One ; 13(4): e0191926, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29617360

RESUMO

CTLA-4 and CD28 exemplify a co-inhibitory and co-stimulatory signaling axis that dynamically sculpts the interaction of antigen-specific T cells with antigen-presenting cells. Anti-CTLA-4 antibodies enhance tumor-specific immunity through a variety of mechanisms including: blockade of CD80 or CD86 binding to CTLA-4, repressing regulatory T cell function and selective elimination of intratumoral regulatory T cells via an Fcγ receptor-dependent mechanism. AGEN1884 is a novel IgG1 antibody targeting CTLA-4. It potently enhanced antigen-specific T cell responsiveness that could be potentiated in combination with other immunomodulatory antibodies. AGEN1884 was well-tolerated in non-human primates and enhanced vaccine-mediated antigen-specific immunity. AGEN1884 combined effectively with PD-1 blockade to elicit a T cell proliferative response in the periphery. Interestingly, an IgG2 variant of AGEN1884 revealed distinct functional differences that may have implications for optimal dosing regimens in patients. Taken together, the pharmacological properties of AGEN1884 support its clinical investigation as a single therapeutic and combination agent.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antineoplásicos Imunológicos/farmacologia , Antígeno CTLA-4/imunologia , Imunoglobulina G/farmacologia , Neoplasias/terapia , Adjuvantes Imunológicos/química , Adjuvantes Imunológicos/farmacocinética , Adjuvantes Imunológicos/toxicidade , Sequência de Aminoácidos , Animais , Formação de Anticorpos/efeitos dos fármacos , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/farmacocinética , Antineoplásicos Imunológicos/toxicidade , Células CHO , Antígeno CTLA-4/antagonistas & inibidores , Vacinas Anticâncer/farmacologia , Células Cultivadas , Cricetulus , Mapeamento de Epitopos , Humanos , Imunidade Celular/efeitos dos fármacos , Imunoglobulina G/química , Imunoglobulina G/toxicidade , Ativação Linfocitária/efeitos dos fármacos , Macaca fascicularis , Modelos Moleculares , Neoplasias/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
4.
Hum Antibodies ; 25(3-4): 87-109, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28085016

RESUMO

Co-stimulatory tumor necrosis factor receptors (TNFRs) can sculpt the responsiveness of T cells recognizing tumor-associated antigens. For this reason, agonist antibodies targeting CD137, CD357, CD134 and CD27 have received considerable attention for their therapeutic utility in enhancing anti-tumor immune responses, particularly in combination with other immuno-modulatory antibodies targeting co-inhibitory pathways in T cells. The design of therapeutic antibodies that optimally engage and activate co-stimulatory TNFRs presents an important challenge of how to promote effective anti-tumor immunity while avoiding serious immune-related adverse events. Here we review our current understanding of the expression, signaling and structural features of CD137, CD357, CD134 and CD27, and how this may inform the design of pharmacologically active immuno-modulatory antibodies targeting these receptors. This includes the integration of our emerging knowledge of the role of Fcγ receptors (FcγRs) in facilitating antibody-mediated receptor clustering and forward signaling, as well as promoting immune effector cell-mediated activities. Finally, we bring our current preclinical and clinical knowledge of co-stimulatory TNFR antibodies into the context of opportunities for next generation molecules with improved pharmacologic properties.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Receptores de IgG/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Animais , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Regulação da Expressão Gênica , Humanos , Imunidade Celular/efeitos dos fármacos , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/patologia , Receptores de IgG/agonistas , Receptores de IgG/genética , Receptores do Fator de Necrose Tumoral/agonistas , Receptores do Fator de Necrose Tumoral/genética , Transdução de Sinais , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/patologia
6.
J Immunol ; 194(3): 878-82, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25548231

RESUMO

CD4(+) regulatory T cells (Tregs) are critical for maintaining self-tolerance and function to prevent autoimmune disease. High densities of intratumoral Tregs are generally associated with poor patient prognosis, a correlation attributed to their broad immune-suppressive features. Two major populations of Tregs have been defined, thymically derived natural Tregs (nTregs) and peripherally induced Tregs (iTregs). However, the relative contribution of nTregs versus iTregs to the intratumoral Treg compartment remains controversial. Demarcating the proportion of nTregs versus iTregs has important implications in the design of therapeutic strategies to overcome their antagonistic effects on antitumor immune responses. We used epigenetic, phenotypic, and functional parameters to evaluate the composition of nTregs versus iTregs isolated from mouse tumor models and primary human tumors. Our findings failed to find evidence for extensive intratumoral iTreg induction. Rather, we identified a population of Foxp3-stable nTregs in tumors from mice and humans.


Assuntos
Epigênese Genética , Fatores de Transcrição Forkhead/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias/genética , Neoplasias/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Antígenos de Superfície/metabolismo , Linhagem Celular Tumoral , Ilhas de CpG , Metilação de DNA , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/metabolismo , Humanos , Imunofenotipagem , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Camundongos Transgênicos , Neoplasias/metabolismo , Fator de Crescimento Transformador beta/metabolismo
7.
Immunol Cell Biol ; 92(6): 475-80, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24732076

RESUMO

Antibodies targeting checkpoint inhibitors or co-stimulatory receptors on T cells have shown significant antitumor efficacy in preclinical and clinical studies. In mouse tumor models, engagement of activating Fcγ receptor (FcγR)-expressing immune cells was recently shown to be required for the tumoricidal activity of antibodies recognizing the tumor necrosis factor superfamily receptor (TNFR) GITR (CD357) and CTLA-4 (CD152). In particular, activating FcγRs facilitated the selective elimination of intratumoral T-cell populations. However, it remains unclear whether FcγRs contribute to the antitumor efficacy of other immunomodulatory antibodies. Here, we explored the mechanism of antitumor activity mediated by an agonistic antibody (clone OX86) to the co-stimulatory TNFR OX40 (CD134). OX40 was highly expressed by intratumoral T cells, particularly those of the FoxP3(+) regulatory T-cell (Treg) lineage. OX86 administration resulted in the depletion of intratumoral regulatory T cells in an activating FcγR-dependent manner, which correlated with tumor regression. Together with previous data from our group and others, these findings support a mechanism whereby antibodies targeting antigens highly expressed by intratumoral T cells can mediate their elimination by FcγR-expressing immune cells, and facilitate subsequent antitumor immunity.


Assuntos
Neoplasias Experimentais/imunologia , Receptores de IgG/imunologia , Receptores OX40/imunologia , Linfócitos T Reguladores/imunologia , Animais , Antígeno CTLA-4/genética , Antígeno CTLA-4/imunologia , Proteína Relacionada a TNFR Induzida por Glucocorticoide/genética , Proteína Relacionada a TNFR Induzida por Glucocorticoide/imunologia , Depleção Linfocítica , Camundongos , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Receptores de IgG/genética , Receptores OX40/genética , Linfócitos T Reguladores/patologia
8.
J Immunol ; 192(7): 3259-68, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24610009

RESUMO

Adjuvants are an essential component of modern vaccines and used for their ability to elicit immunity to coadministered Ags. Many adjuvants in clinical development are particulates, but how they drive innate and adaptive immune responses remains poorly understood. Studies have shown that a number of vaccine adjuvants activate inflammasome pathways in isolated APCs. However, the contribution of inflammasome activation to vaccine-mediated immunity in vivo remains controversial. In this study, we evaluated immune cell responses to the ISCOMATRIX adjuvant (IMX) in mice. Like other particulate vaccine adjuvants, IMX potently activated the NALP-3-ASC-Caspase-1 inflammasome in APCs, leading to IL-1ß and IL-18 production. The IL-18R pathway, but not IL-1R, was required for early innate and subsequent cellular immune responses to a model IMX vaccine. APCs directly exposed to IMX underwent an endosome-mediated cell-death response, which we propose initiates inflammatory events locally at the injection site. Importantly, both inflammasome-related and -unrelated pathways contributed to IL-18 dependence in vivo following IMX administration. TNF-α provided a physiological priming signal for inflammasome-dependent IL-18 production by APCs, which correlated with reduced vaccine-mediated immune cell responses in TNF-α- or TNFR-deficient mice. Taken together, our findings highlight an important disconnect between the mechanisms of vaccine adjuvant action in vitro versus in vivo.


Assuntos
Colesterol/imunologia , Imunidade/imunologia , Inflamassomos/imunologia , Interleucina-18/imunologia , Fosfolipídeos/imunologia , Saponinas/imunologia , Trifosfato de Adenosina/imunologia , Trifosfato de Adenosina/metabolismo , Adjuvantes Imunológicos/farmacologia , Animais , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Colesterol/farmacologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Combinação de Medicamentos , Humanos , Imunidade/efeitos dos fármacos , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Lisossomos/efeitos dos fármacos , Lisossomos/imunologia , Lisossomos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Fosfolipídeos/farmacologia , Receptores do Fator de Necrose Tumoral/deficiência , Receptores do Fator de Necrose Tumoral/genética , Receptores do Fator de Necrose Tumoral/imunologia , Saponinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
9.
Immunol Cell Biol ; 91(7): 451-60, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23817579

RESUMO

Toll-like receptors (TLRs) enable metazoans to mount effective innate immune responses to microbial and viral pathogens, as well as to endogenous host-derived ligands. It is understood that genetic background of the host can influence TLR responsiveness, altering susceptibility to pathogen infection, autoimmunity and cancer. Macrophage stimulatory protein (MSP), which activates the receptor tyrosine kinase recepteur d'origine nantais (RON), promotes key macrophage functions such as motility and phagocytic activity. MSP also acts via RON to modulate signaling by TLR4, which recognizes a range of pathogen or endogenous host-derived molecules. Here, we show that RON exerts divergent control over TLR4 activity in macrophages from different mouse genetic backgrounds. RON potently modulated the TLR4 response in macrophages from M2-prone FVB mice, as compared with M1-skewed C57Bl6 mice. Moreover, global expression analysis revealed that RON suppresses the TLR4-dependent type-I interferon gene signature only in FVB macrophages. This leads to attenuated production of the potent inflammatory mediator, tumor necrosis factor-α. Eliminating RON kinase activity markedly decreased carcinogen-mediated tumorigenesis in M2/Th2-biased FVB mice. We propose that host genetic background influences RON function, thereby contributing to the variability in TLR4 responsiveness in rodents and, potentially, in humans. These findings provide novel insight into the complex interplay between genetic context and immune function.


Assuntos
Fibrossarcoma/imunologia , Macrófagos Peritoneais/imunologia , Papiloma/imunologia , Receptores Proteína Tirosina Quinases/metabolismo , Neoplasias Cutâneas/imunologia , Receptor 4 Toll-Like/imunologia , 9,10-Dimetil-1,2-benzantraceno/administração & dosagem , Animais , Carcinogênese , Movimento Celular/efeitos dos fármacos , Fibrossarcoma/induzido quimicamente , Fibrossarcoma/genética , Genótipo , Fator de Crescimento de Hepatócito/metabolismo , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Metilcolantreno/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Papiloma/induzido quimicamente , Papiloma/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/genética , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/genética , Equilíbrio Th1-Th2 , Ativação Transcricional/genética , Transcriptoma
10.
J Exp Med ; 210(9): 1685-93, 2013 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-23897982

RESUMO

Fc γ receptor (FcγR) coengagement can facilitate antibody-mediated receptor activation in target cells. In particular, agonistic antibodies that target tumor necrosis factor receptor (TNFR) family members have shown dependence on expression of the inhibitory FcγR, FcγRIIB. It remains unclear if engagement of FcγRIIB also extends to the activities of antibodies targeting immunoregulatory TNFRs expressed by T cells. We have explored the requirement for activating and inhibitory FcγRs for the antitumor effects of antibodies targeting the TNFR glucocorticoid-induced TNFR-related protein (GITR; TNFRSF18; CD357) expressed on activated and regulatory T cells (T reg cells). We found that although FcγRIIB was dispensable for the in vivo efficacy of anti-GITR antibodies, in contrast, activating FcγRs were essential. Surprisingly, the dependence on activating FcγRs extended to an antibody targeting the non-TNFR receptor CTLA-4 (CD152) that acts as a negative regulator of T cell immunity. We define a common mechanism that correlated with tumor efficacy, whereby antibodies that coengaged activating FcγRs expressed by tumor-associated leukocytes facilitated the selective elimination of intratumoral T cell populations, particularly T reg cells. These findings may have broad implications for antibody engineering efforts aimed at enhancing the therapeutic activity of immunomodulatory antibodies.


Assuntos
Anticorpos Antineoplásicos/farmacologia , Antineoplásicos/farmacologia , Proteína Relacionada a TNFR Induzida por Glucocorticoide/antagonistas & inibidores , Fatores Imunológicos/farmacologia , Neoplasias/imunologia , Receptores de IgG/metabolismo , Animais , Antígeno CTLA-4/metabolismo , Feminino , Proteína Relacionada a TNFR Induzida por Glucocorticoide/metabolismo , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/patologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo
11.
Cancer Cell ; 22(1): 80-90, 2012 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-22789540

RESUMO

The proapoptotic death receptor DR5 has been studied extensively in cancer cells, but its action in the tumor microenvironment is not well defined. Here, we uncover a role for DR5 signaling in tumor endothelial cells (ECs). We detected DR5 expression in ECs within tumors but not normal tissues. Treatment of tumor-bearing mice with an oligomeric form of the DR5 ligand Apo2L/TRAIL induced apoptosis in tumor ECs, collapsing blood vessels and reducing tumor growth: Vascular disruption and antitumor activity required DR5 expression on tumor ECs but not malignant cells. These results establish a therapeutic paradigm for proapoptotic receptor agonists as selective tumor vascular disruption agents, providing an alternative, perhaps complementary, strategy to their use as activators of apoptosis in malignant cells.


Assuntos
Apoptose , Divisão Celular , Endotélio Vascular/metabolismo , Neoplasias/irrigação sanguínea , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Humanos , Camundongos , Neoplasias/patologia
12.
Immunol Cell Biol ; 90(5): 540-52, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-21894173

RESUMO

Generating a cytotoxic CD8(+) T-cell response that can eradicate malignant cells is the primary objective of cancer vaccine strategies. In this study we have characterized the innate and adaptive immune response to the ISCOMATRIX adjuvant, and the ability of vaccine antigens formulated with this adjuvant to promote antitumor immunity. ISCOMATRIX adjuvant led to a rapid innate immune cell response at the injection site, followed by the activation of natural killer and dendritic cells (DC) in regional draining lymph nodes. Strikingly, major histocompatibility complex (MHC) class I cross-presentation by CD8α(+) and CD8α(-) DCs was enhanced by up to 100-fold when antigen was formulated with ISCOMATRIX adjuvant. These coordinated features enabled efficient CD8(+) T-cell cross-priming, which exhibited prophylactic and therapeutic tumoricidal activity. The therapeutic efficacy of an ISCOMATRIX vaccine was further improved when co-administered with an anti-CD40 agonist antibody, suggesting that ISCOMATRIX-based vaccines may combine favorably with other immune modifiers in clinical development to treat cancer. Finally, we identified a requirement for the myeloid differentiation primary response gene 88 (MyD88) adapter protein for both innate and adaptive immune responses to ISCOMATRIX vaccines in vivo. Taken together, our findings support the utility of the ISCOMATRIX adjuvant for use in the development of novel vaccines, particularly those requiring strong CD8(+) T-cell immune responses, such as therapeutic cancer vaccines.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Colesterol/imunologia , Fosfolipídeos/imunologia , Saponinas/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Antígenos de Neoplasias/imunologia , Antígenos CD40/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Vacinas Anticâncer/administração & dosagem , Colesterol/administração & dosagem , Apresentação Cruzada/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Combinação de Medicamentos , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Ovalbumina/imunologia , Fosfolipídeos/administração & dosagem , Receptor Cross-Talk/efeitos dos fármacos , Saponinas/administração & dosagem , Transdução de Sinais/efeitos dos fármacos
13.
Cancer Cell ; 19(1): 101-13, 2011 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-21251615

RESUMO

Antibodies to cell-surface antigens trigger activatory Fcγ receptor (FcγR)-mediated retrograde signals in leukocytes to control immune effector functions. Here, we uncover an FcγR mechanism that drives antibody-dependent forward signaling in target cells. Agonistic antibodies to death receptor 5 (DR5) induce cancer-cell apoptosis and are in clinical trials; however, their mechanism of action in vivo is not fully defined. Interaction of the DR5-agonistic antibody drozitumab with leukocyte FcγRs promoted DR5-mediated tumor-cell apoptosis. Whereas the anti-CD20 antibody rituximab required activatory FcγRs for tumoricidal function, drozitumab was effective in the context of either activatory or inhibitory FcγRs. A CD40-agonistic antibody required similar FcγR interactions to stimulate nuclear factor-κB activity in B cells. Thus, FcγRs can drive antibody-mediated receptor signaling in target cells.


Assuntos
Anticorpos Monoclonais/metabolismo , Neoplasias/metabolismo , Receptores de IgG/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Transdução de Sinais/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Apoptose/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Antígenos CD40/agonistas , Antígenos CD40/imunologia , Linhagem Celular Tumoral , Feminino , Células HCT116 , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Imunoglobulina G/farmacologia , Células Matadoras Naturais/imunologia , Leucócitos/imunologia , Leucócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Mutação/imunologia , Células Mieloides/imunologia , NF-kappa B/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Polimorfismo de Nucleotídeo Único/genética , Polimorfismo de Nucleotídeo Único/imunologia , Ligação Proteica/genética , Ligação Proteica/imunologia , Agregação de Receptores/imunologia , Receptores de IgG/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Curr Opin Cell Biol ; 22(6): 837-44, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20813513

RESUMO

Proapoptotic receptor agonists (PARAs) targeting death receptors (DRs) 4 and 5 hold promise for cancer therapy based on their selective ability to kill malignant versus healthy cells. Emerging clinical results have confirmed that DR4/5 PARAs are relatively well-tolerated and suitable for further investigation. Given that some cancer cell lines and models are not sensitive to PARAs, it is important to develop strategies to identify what specific types of tumor cells may be most responsive to PARA-based therapy and how to overcome apoptosis resistance mechanisms in tumors. Here we review the molecular and biological determinants of responsiveness to PARAs in cancer cells, and discuss the potential for predictive biomarkers and drug combination strategies to maximize the anti-tumor activity of these agents.


Assuntos
Apoptose/fisiologia , Neoplasias/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Transdução de Sinais/fisiologia , Animais , Membrana Celular/metabolismo , Humanos , Mitocôndrias/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
15.
Nat Immunol ; 10(4): 348-55, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19295631

RESUMO

Death receptors (DRs) are members of the tumor necrosis factor receptor superfamily that possess a cytoplasmic death domain (DD). DRs regulate important operational and homeostatic aspects of the immune system. They transmit signals through apical protein complexes, which are nucleated by the DD adaptors FADD and TRADD, to control cellular outcomes that range from apoptosis to gene activation. FADD and TRADD also nucleate several distal signaling complexes, which mediate cross-talk between distinct DR signaling pathways. Moreover, together with other DR signal transducers, FADD and TRADD participate in functional complexes assembled by certain non-DR immune cell receptors, such as pattern-recognition receptors. Thus, DR signal transducers may provide important nodes of coordination in immune signaling networks.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteína de Domínio de Morte Associada a Fas/fisiologia , Receptores de Morte Celular/fisiologia , Transdução de Sinais/fisiologia , Proteína de Domínio de Morte Associada a Receptor de TNF/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Apoptose/fisiologia , Proteína de Domínio de Morte Associada a Fas/imunologia , Humanos , Imunidade Ativa , Imunidade Inata , Receptores de Morte Celular/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/fisiologia , Proteína de Domínio de Morte Associada a Receptor de TNF/imunologia
16.
Eur J Immunol ; 36(2): 327-35, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16402408

RESUMO

While naive CD8(+) T cells have been shown to require bone marrow-derived dendritic cells (DC) to initiate immunity, such a requirement for memory CD8(+) T cells has had limited assessment. By generating bone marrow chimeras that express the appropriate antigen-presenting molecules on either radiation-sensitive bone marrow-derived or radiation-resistant non-bone marrow-derived compartments, we showed that both primary and secondary immune responses to influenza virus infection of the lung were initiated in the draining LN. This required cells of bone marrow origin, most likely DC, for optimal expansion within the secondary lymphoid compartment. This was similarly the case with HSV-1 infection of the skin. As Langerhans cells are radioresistant, unlike other DC populations, these studies also demonstrate that the radiosensitive DC responsible for secondary expansion of HSV-specific memory are not Langerhans cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Herpes Simples/imunologia , Memória Imunológica/imunologia , Pneumopatias/imunologia , Infecções por Orthomyxoviridae/imunologia , Orthomyxoviridae/imunologia , Simplexvirus/imunologia , Animais , Células da Medula Óssea/imunologia , Transplante de Medula Óssea , Feminino , Herpes Simples/genética , Memória Imunológica/genética , Memória Imunológica/efeitos da radiação , Células de Langerhans/imunologia , Pneumopatias/genética , Linfonodos/imunologia , Masculino , Camundongos , Camundongos Knockout , Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/genética , Quimeras de Transplante , Irradiação Corporal Total
17.
J Immunol ; 174(11): 6592-7, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15905497

RESUMO

We demonstrate that functional and phenotypic equivalents of mouse splenic CD8(+) and CD8(-) conventional dendritic cell (cDC) subsets can be generated in vitro when bone marrow is cultured with fms-like tyrosine kinase 3 (flt3) ligand. In addition to CD45RA(high) plasmacytoid DC, two distinct CD24(high) and CD11b(high) cDC subsets were present, and these subsets showed equivalent properties to splenic CD8(+) and CD8(-) cDC, respectively, in the following: 1) surface expression of CD11b, CD24, and signal regulatory protein-alpha; 2) developmental dependence on, and mRNA expression of, IFN regulatory factor-8; 3) mRNA expression of TLRs and chemokine receptors; 4) production of IL-12 p40/70, IFN-alpha, MIP-1alpha, and RANTES in response to TLR ligands; 5) expression of cystatin C; and 6) cross-presentation of exogenous Ag to CD8 T cells. Furthermore, despite lacking surface CD8 expression, the CD24(high) subset contained CD8 mRNA and up-regulated surface expression when transferred into mice. This culture system allows access to bona fide counterparts of the splenic DC subsets.


Assuntos
Células da Medula Óssea/imunologia , Antígenos CD8/biossíntese , Diferenciação Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Baço/imunologia , Animais , Apresentação de Antígeno/genética , Apresentação de Antígeno/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Antígenos CD4/biossíntese , Antígenos CD4/genética , Antígenos CD8/genética , Diferenciação Celular/genética , Células Cultivadas , Quimiocinas/biossíntese , Apresentação Cruzada/genética , Apresentação Cruzada/imunologia , Cistatina C , Cistatinas/biossíntese , Citocinas/biossíntese , Células Dendríticas/metabolismo , Imunofenotipagem , Fatores Reguladores de Interferon , Ligantes , Glicoproteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores de Superfície Celular/biossíntese , Receptores de Quimiocinas/biossíntese , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Baço/citologia , Baço/metabolismo , Receptores Toll-Like , Tirosina Quinase 3 Semelhante a fms
18.
Immunol Rev ; 199: 9-26, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15233723

RESUMO

Cross-presentation involves the uptake and processing of exogenous antigens within the major histocompatibility complex (MHC) class I pathway. This process is primarily performed by dendritic cells (DCs), which are not a single cell type but may be divided into several distinct subsets. Those expressing CD8alpha together with CD205, found primarily in the T-cell areas of the spleen and lymph nodes, are the major subset responsible for cross-presenting cellular antigens. This ability is likely to be important for the generation of cytotoxic T-cell immunity to a variety of antigens, particularly those associated with viral infection, tumorigenesis, and DNA vaccination. At present, it is unclear whether the CD8alpha-expressing DC subset captures antigen directly from target cells or obtains it indirectly from intermediary DCs that traffic from peripheral sites. In this review, we examine the molecular basis for cross-presentation, discuss the role of DC subsets, and examine the contribution of this process to immunity, with some emphasis on DNA vaccination.


Assuntos
Antígenos/imunologia , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Vacinas de DNA/imunologia , Animais , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Células Dendríticas/classificação , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Vacinas de DNA/genética
19.
J Immunol ; 171(10): 5003-11, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14607896

RESUMO

Dendritic cells (DC) undergo complex developmental changes during maturation. The MHC class II (MHC II) molecules of immature DC accumulate in intracellular compartments, but are expressed at high levels on the plasma membrane upon DC maturation. It has been proposed that the cysteine protease inhibitor cystatin C (CyC) plays a pivotal role in the control of this process by regulating the activity of cathepsin S, a protease involved in removal of the MHC II chaperone Ii, and hence in the formation of MHC II-peptide complexes. We show that CyC is differentially expressed by mouse DC populations. CD8(+) DC, but not CD4(+) or CD4(-)CD8(-) DC, synthesize CyC, which accumulates in MHC II(+)Lamp(+) compartments. However, Ii processing and MHC II peptide loading proceeded similarly in all three DC populations. We then analyzed MHC II localization and Ag presentation in CD8(+) DC, bone marrow-derived DC, and spleen-derived DC lines, from CyC-deficient mice. The absence of CyC did not affect the expression, the subcellular distribution, or the formation of peptide-loaded MHC II complexes in any of these DC types, nor the efficiency of presentation of exogenous Ags. Therefore, CyC is neither necessary nor sufficient to control MHC II expression and Ag presentation in DC. Our results also show that CyC expression can differ markedly between closely related cell types, suggesting the existence of hitherto unrecognized mechanisms of control of CyC expression.


Assuntos
Apresentação de Antígeno/fisiologia , Cistatinas/biossíntese , Cistatinas/fisiologia , Inibidores de Cisteína Proteinase/biossíntese , Inibidores de Cisteína Proteinase/fisiologia , Células Dendríticas/enzimologia , Células Dendríticas/imunologia , Animais , Antígenos de Diferenciação de Linfócitos B/biossíntese , Antígenos de Diferenciação de Linfócitos B/metabolismo , Antígenos CD4/biossíntese , Antígenos CD8/biossíntese , Catepsinas/deficiência , Catepsinas/genética , Catepsinas/fisiologia , Células Cultivadas , Cistatina C , Cistatinas/deficiência , Cistatinas/genética , Células Dendríticas/metabolismo , Antígenos de Histocompatibilidade Classe II/biossíntese , Antígenos de Histocompatibilidade Classe II/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/metabolismo , Processamento de Proteína Pós-Traducional/imunologia , Baço/citologia , Baço/imunologia , Baço/metabolismo , Frações Subcelulares/imunologia , Frações Subcelulares/metabolismo
20.
J Immunol ; 170(9): 4437-40, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12707318

RESUMO

CTL play a major role in immunity to HSV type 1, but little is known about the priming process. In this study, we have examined the class I-restricted presentation of an immunodominant determinant from HSV-1 glycoprotein B after footpad infection. We have found that the only cell types capable of presenting this determinant in draining popliteal lymph nodes within the first 3 days after infection are the CD11c(+)CD8alpha(+)CD45RA(-) dendritic cells. Given that such class I-restricted presentation is essential for CTL priming, this implies that these conventional CD8alpha(+) dendritic cells are the key subset involved in CTL immunity to this virus.


Assuntos
Antígenos CD8/biossíntese , Citotoxicidade Imunológica/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Herpes Simples/imunologia , Herpesvirus Humano 1/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Células Apresentadoras de Antígenos/virologia , Células Dendríticas/virologia , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Herpes Simples/virologia , Membro Posterior , Antígenos de Histocompatibilidade Classe I/metabolismo , Hibridomas , Imunidade Ativa , Epitopos Imunodominantes/imunologia , Epitopos Imunodominantes/metabolismo , Injeções Subcutâneas , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Envelope Viral/imunologia , Proteínas do Envelope Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA