Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Immunity ; 43(5): 835-7, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26588774

RESUMO

How inflammatory caspases trigger pyroptotic cell death is mostly unexplained. In this issue of Immunity, Núñez and colleagues report that caspase-11 cleaves the transmembrane channel pannexin-1, causing an efflux of cellular ATP that promotes a P2X7 receptor-dependent pyroptosis.


Assuntos
Caspases/metabolismo , Conexinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Piroptose/fisiologia , Receptores Purinérgicos P2X7/metabolismo , Choque Séptico/metabolismo , Animais , Humanos
2.
Blood ; 129(17): 2420-2428, 2017 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-28167662

RESUMO

The endoplasmic reticulum kinase inositol-requiring enzyme 1 (IRE1) and its downstream target X-box-binding protein 1 (XBP1) drive B-cell differentiation toward plasma cells and have been shown to contribute to multiple myeloma development; yet, little is known of the role of this pathway in diffuse large B-cell lymphoma (DLBCL). Here, we show that in the germinal center B-cell-like (GCB) DLBCL subtype, IRE1 expression is reduced to a level that prevents XBP1 activation. Gene expression profiles indicated that, in GCB DLBCL cancer samples, expression of IRE1 messenger RNA was inversely correlated with the levels and activity of the epigenetic repressor, histone methyltransferase enhancer of zeste homolog 2 (EZH2). Correspondingly, in GCB-derived cell lines, the IRE1 promoter carried increased levels of the repressive epigenetic mark histone 3 lysine 27 trimethylation. Pharmacological inhibition of EZH2 erased those marks and restored IRE1 expression and function in vitro and in vivo. Moreover, reconstitution of the IRE1-signaling pathway, by expression of the XBP1-active form, compromised GCB DLBCL tumor growth in a mouse xenograft cancer model. These findings indicate that IRE1-XBP1 downregulation distinguishes GCB DLBCL from other DLBCL subtypes and contributes to tumor growth.


Assuntos
Linfócitos B/imunologia , Endorribonucleases/genética , Regulação Neoplásica da Expressão Gênica , Centro Germinativo/imunologia , Linfoma Difuso de Grandes Células B/imunologia , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , Proteína 1 de Ligação a X-Box/genética , Animais , Linfócitos B/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Endorribonucleases/antagonistas & inibidores , Endorribonucleases/imunologia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/imunologia , Epigênese Genética , Centro Germinativo/patologia , Histonas/genética , Histonas/imunologia , Humanos , Indazóis/farmacologia , Leupeptinas/farmacologia , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/patologia , Linfoma Difuso de Grandes Células B/terapia , Camundongos , Camundongos da Linhagem 129 , Plasmócitos/imunologia , Plasmócitos/patologia , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/imunologia , Piridonas/farmacologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/imunologia , Transdução de Sinais , Proteína 1 de Ligação a X-Box/antagonistas & inibidores , Proteína 1 de Ligação a X-Box/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Proc Natl Acad Sci U S A ; 113(2): E117-26, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26715744

RESUMO

Inhibitors of the HIV aspartyl protease [HIV protease inhibitors (HIV-PIs)] are the cornerstone of treatment for HIV. Beyond their well-defined antiretroviral activity, these drugs have additional effects that modulate cell viability and homeostasis. However, little is known about the virus-independent pathways engaged by these molecules. Here we show that the HIV-PI Nelfinavir decreases translation rates and promotes a transcriptional program characteristic of the integrated stress response (ISR). Mice treated with Nelfinavir display hallmarks of this stress response in the liver, including α subunit of translation initiation factor 2 (eIF2α) phosphorylation, activating transcription factor-4 (ATF4) induction, and increased expression of known downstream targets. Mechanistically, Nelfinavir-mediated ISR bypassed direct activation of the eIF2α stress kinases and instead relied on the inhibition of the constitutive eIF2α dephosphorylation and down-regulation of the phophatase cofactor CReP (Constitutive Repressor of eIF2α Phosphorylation; also known as PPP1R15B). These findings demonstrate that the modulation of eIF2α-specific phosphatase cofactor activity can be a rheostat of cellular homeostasis that initiates a functional ISR and suggest that the HIV-PIs could be repositioned as therapeutics in human diseases to modulate translation rates and stress responses.


Assuntos
Fator de Iniciação 2 em Eucariotos/metabolismo , Inibidores da Protease de HIV/farmacologia , Protease de HIV/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Animais , Células HeLa , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Proteínas de Membrana/deficiência , Proteínas de Membrana/metabolismo , Camundongos , Nelfinavir/farmacologia , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 1/metabolismo , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/metabolismo , Transcrição Gênica/efeitos dos fármacos , Resposta a Proteínas não Dobradas/efeitos dos fármacos , eIF-2 Quinase/metabolismo
4.
Proc Natl Acad Sci U S A ; 113(32): E4671-80, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27462105

RESUMO

Inflammasomes are critical sensors that convey cellular stress and pathogen presence to the immune system by activating inflammatory caspases and cytokines such as IL-1ß. The nature of endogenous stress signals that activate inflammasomes remains unclear. Here we show that an inhibitor of the HIV aspartyl protease, Nelfinavir, triggers inflammasome formation and elicits an IL-1R-dependent inflammation in mice. We found that Nelfinavir impaired the maturation of lamin A, a structural component of the nuclear envelope, thereby promoting the release of DNA in the cytosol. Moreover, deficiency of the cytosolic DNA-sensor AIM2 impaired Nelfinavir-mediated inflammasome activation. These findings identify a pharmacologic activator of inflammasome and demonstrate the role of AIM2 in detecting endogenous DNA release upon perturbation of nuclear envelope integrity.


Assuntos
Inflamassomos/efeitos dos fármacos , Nelfinavir/farmacologia , Membrana Nuclear/efeitos dos fármacos , Animais , Proteínas Adaptadoras de Sinalização CARD/fisiologia , Caspase 1/metabolismo , DNA/metabolismo , Inflamassomos/fisiologia , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Membrana Nuclear/fisiologia , Receptores de Interleucina-1/fisiologia
5.
EMBO Rep ; 17(10): 1471-1484, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27572820

RESUMO

Activation of the elongation factor 2 kinase (eEF2K) leads to the phosphorylation and inhibition of the elongation factor eEF2, reducing mRNA translation rates. Emerging evidence indicates that the regulation of factors involved in protein synthesis may be critical for controlling diverse biological processes including cancer progression. Here we show that inhibitors of the HIV aspartyl protease (HIV-PIs), nelfinavir in particular, trigger a robust activation of eEF2K leading to the phosphorylation of eEF2. Beyond its anti-viral effects, nelfinavir has antitumoral activity and promotes cell death. We show that nelfinavir-resistant cells specifically evade eEF2 inhibition. Decreased cell viability induced by nelfinavir is impaired in cells lacking eEF2K. Moreover, nelfinavir-mediated anti-tumoral activity is severely compromised in eEF2K-deficient engrafted tumors in vivo Our findings imply that exacerbated activation of eEF2K is detrimental for tumor survival and describe a mechanism explaining the anti-tumoral properties of HIV-PIs.


Assuntos
Quinase do Fator 2 de Elongação/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Modelos Animais de Doenças , Progressão da Doença , Relação Dose-Resposta a Droga , Resistência a Medicamentos/genética , Quinase do Fator 2 de Elongação/genética , Feminino , Expressão Gênica , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Complexos Multiproteicos/metabolismo , Nelfinavir/química , Nelfinavir/farmacologia , Neoplasias/genética , Fator 2 de Elongação de Peptídeos/metabolismo , Fosforilação , Biossíntese de Proteínas , Serina-Treonina Quinases TOR/metabolismo , Carga Tumoral
6.
J Immunol ; 196(7): 2939-46, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26944927

RESUMO

NLRC5, a member of the NOD-like receptor (NLR) protein family, has recently been characterized as the master transcriptional regulator of MHCI molecules in lymphocytes, in which it is highly expressed. However, its role in activated dendritic cells (DCs), which are instrumental to initiate T cell responses, remained elusive. We show in this study that, following stimulation of DCs with inflammatory stimuli, not only did NLRC5 level increase, but also its importance in directing MHCI transcription. Despite markedly reduced mRNA and intracellular H2-K levels, we unexpectedly observed nearly normal H2-K surface display in Nlrc5(-/-) DCs. Importantly, this discrepancy between a strong intracellular and a mild surface defect in H2-K levels was observed also in DCs with H2-K transcription defects independent of Nlrc5. Hence, alongside with demonstrating the importance of NLRC5 in MHCI transcription in activated DCs, we uncover a general mechanism counteracting low MHCI surface expression. In agreement with the decreased amount of neosynthesized MHCI, Nlrc5(-/-) DCs exhibited a defective capacity to display endogenous Ags. However, neither T cell priming by endogenous Ags nor cross-priming ability was substantially affected in activated Nlrc5(-/-) DCs. Altogether, these data show that Nlrc5 deficiency, despite significantly affecting MHCI transcription and Ag display, is not sufficient to hinder T cell activation, underlining the robustness of the T cell priming process by activated DCs.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Animais , Apresentação de Antígeno/imunologia , Linhagem Celular , Membrana Celular/metabolismo , Apresentação Cruzada/imunologia , Regulação da Expressão Gênica , Ativação Linfocitária/genética , Camundongos , Camundongos Knockout , Linfócitos T/metabolismo , Transcrição Gênica
7.
Proc Natl Acad Sci U S A ; 112(50): 15408-13, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26607445

RESUMO

Spontaneous CD8 T-cell responses occur in growing tumors but are usually poorly effective. Understanding the molecular and cellular mechanisms that drive these responses is of major interest as they could be exploited to generate a more efficacious antitumor immunity. As such, stimulator of IFN genes (STING), an adaptor molecule involved in cytosolic DNA sensing, is required for the induction of antitumor CD8 T responses in mouse models of cancer. Here, we find that enforced activation of STING by intratumoral injection of cyclic dinucleotide GMP-AMP (cGAMP), potently enhanced antitumor CD8 T responses leading to growth control of injected and contralateral tumors in mouse models of melanoma and colon cancer. The ability of cGAMP to trigger antitumor immunity was further enhanced by the blockade of both PD1 and CTLA4. The STING-dependent antitumor immunity, either induced spontaneously in growing tumors or induced by intratumoral cGAMP injection was dependent on type I IFNs produced in the tumor microenvironment. In response to cGAMP injection, both in the mouse melanoma model and an ex vivo model of cultured human melanoma explants, the principal source of type I IFN was not dendritic cells, but instead endothelial cells. Similarly, endothelial cells but not dendritic cells were found to be the principal source of spontaneously induced type I IFNs in growing tumors. These data identify an unexpected role of the tumor vasculature in the initiation of CD8 T-cell antitumor immunity and demonstrate that tumor endothelial cells can be targeted for immunotherapy of melanoma.


Assuntos
Células Endoteliais/metabolismo , Imunidade , Proteínas de Membrana/metabolismo , Neoplasias/imunologia , Neoplasias/terapia , Animais , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígeno CTLA-4/imunologia , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Relação Dose-Resposta Imunológica , Células Endoteliais/efeitos dos fármacos , Injeções Intralesionais , Interferon Tipo I/metabolismo , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Melanoma/imunologia , Melanoma/patologia , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Neoplasias/patologia , Nucleotídeos Cíclicos/administração & dosagem , Nucleotídeos Cíclicos/farmacologia , Receptor de Interferon alfa e beta/metabolismo , Transdução de Sinais/efeitos dos fármacos
8.
Immunol Cell Biol ; 95(9): 753-764, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28559542

RESUMO

Given the heterogeneous nature of antigens, major histocompatibility complex class I (MHC I) intracellular transport intersects with multiple degradation pathways for efficient peptide loading and presentation to cytotoxic T cells. MHC I loading with peptides in the endoplasmic reticulum (ER) is a tightly regulated process, while post-ER intracellular transport is considered to occur by default, leading to peptide-bearing MHC I delivery to the plasma membrane. We show here that MHC I traffic is submitted to a previously uncharacterized sorting step at the trans Golgi network (TGN), dependent on the ubiquitination of its cytoplasmic tail lysine residues. MHC I ubiquitination is mediated by the E3 ligase membrane-associated RING-CH 9 (MARCH9) and allows MHC I access to Syntaxin 6-positive endosomal compartments. We further show that MARCH9 can also target the human MHC I-like lipid antigen-presentation molecule CD1a. MARCH9 expression is modulated by microbial pattern exposure in dendritic cells (DCs), thus revealing the role of this ubiquitin E3 ligase in coordinating MHC I access to endosomes and DC activation for efficient antigen cross-presentation.


Assuntos
Antígenos CD1/metabolismo , Membrana Celular/metabolismo , Células Dendríticas/imunologia , Endossomos/metabolismo , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Rede trans-Golgi/metabolismo , Apresentação de Antígeno , Antígenos CD1/genética , Células Cultivadas , Retículo Endoplasmático/metabolismo , Antígenos HLA/genética , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Proteínas de Membrana , Monócitos/imunologia , Domínios Proteicos/genética , Sinais Direcionadores de Proteínas/genética , Transporte Proteico , Proteínas Qa-SNARE/metabolismo , Ubiquitina-Proteína Ligases , Ubiquitinação
9.
Blood ; 118(3): 609-17, 2011 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-21642595

RESUMO

The brain and dendritic cell (BAD)-associated lysosome-associated membrane protein (LAMP)-like molecule (BAD-LAMP, c20orf103, UNC-46) is a newly identified member of the family of LAMPs. BAD-LAMP expression in the mouse is confined to neurons. We demonstrate here that in humans, BAD-LAMP can specifically be found in the type I IFN-producing plasmacytoid dendritic cells (pDCs). Human BAD-LAMP is localized in the endoplasmic reticulum-Golgi intermediate compartment (ERGIC) of freshly isolated CD123(+) pDCs and is rapidly lost upon activation by unmethylated cytosine-phosphate-guanine (CpG) oligonucleotides. The restricted pattern of BAD-LAMP expression allows for the rapid identification of normal and leukemic human pDCs in tissues and blood.


Assuntos
Biomarcadores/metabolismo , Células Dendríticas/metabolismo , Leucemia/patologia , Proteínas de Membrana Lisossomal/metabolismo , Proteínas de Membrana/metabolismo , Encéfalo/metabolismo , Células Dendríticas/citologia , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Interleucina-3/metabolismo , Subunidade alfa de Receptor de Interleucina-3/metabolismo , Leucemia/metabolismo , Proteínas de Membrana Lisossomal/genética , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oligonucleotídeos/genética , Oligonucleotídeos/metabolismo , Transfecção
10.
Cell Death Dis ; 13(5): 475, 2022 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-35589686

RESUMO

Proteasome inhibitors, such as bortezomib, are first-line therapy against multiple myeloma (MM). Unfortunately, patients frequently become refractory to this treatment. The transcription factor NRF1 has been proposed to initiate an adaptation program that regulates proteasome levels. In the context of proteasome inhibition, the cytosolic protease DDI2 cleaves NRF1 to release an active fragment that translocates to the nucleus to promote the transcription of new proteasome subunits. However, the contribution of the DDI2-NRF1 pathway to bortezomib resistance is poorly understood. Here we show that upon prolonged bortezomib treatment, MM cells become resistant to proteasome inhibition by increasing the expression of DDI2 and consequently activation of NRF1. Furthermore, we found that many MM cells became more sensitive to proteasome impairment in the context of DDI2 deficiency. Mechanistically, we demonstrate that both the protease and the HDD domains of DDI2 are required to activate NRF1. Finally, we show that partial inhibition of the DDI2-protease domain with the antiviral drug nelfinavir increased bortezomib susceptibility in treated MM cells. Altogether, these findings define the DDI2-NRF1 pathway as an essential program contributing to proteasome inhibition responses and identifying DDI2 domains that could be targets of interest in bortezomib-treated MM patients.


Assuntos
Antineoplásicos , Ácido Aspártico Proteases , Mieloma Múltiplo , Antineoplásicos/uso terapêutico , Ácido Aspártico Endopeptidases , Ácido Aspártico Proteases/metabolismo , Bortezomib/farmacologia , Bortezomib/uso terapêutico , Linhagem Celular Tumoral , Humanos , Mieloma Múltiplo/tratamento farmacológico , Inibidores de Proteases/farmacologia , Inibidores de Proteases/uso terapêutico , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Inibidores de Proteassoma/uso terapêutico
11.
iScience ; 25(10): 105227, 2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36248746

RESUMO

DNA-damage inducible 1 homolog 2 (DDI2) is a protease that activates the transcription factor NRF1. Cellular models have shown that this pathway contributes to cell-stress adaptation, for example, on proteasome inhibition. However, DDI2 physiological function is unknown. Ddi2 Knock-out (KO) mice were embryonic lethal. Therefore, we generated liver-specific Ddi2-KO animals and used comprehensive genetic analysis to identify the molecular pathways regulated by DDI2. Here, we demonstrate that DDI2 contributes to metallothionein (MT) expression in mouse and human hepatocytes at basal and upon cadmium (Cd) exposure. This transcriptional program is dependent on DDI2-mediated NRF1 proteolytic maturation. In contrast, NRF1 homolog NRF2 does not contribute to MT production. Mechanistically, we observed that Cd exposure inhibits proteasome activity, resulting in DDI2-mediated NRF1 proteolytic maturation. In line with these findings, DDI2 deficiency sensitizes cells to Cd toxicity. This study identifies a function for DDI2 that links proteasome homeostasis to heavy metal mediated toxicity.

12.
Proc Natl Acad Sci U S A ; 105(9): 3491-6, 2008 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-18305173

RESUMO

In response to Toll-like receptor ligands, dendritic cells (DCs) dramatically enhance their antigen presentation capacity by stabilizing at the cell-surface MHC II molecules. We demonstrate here that, in human monocyte-derived DCs, the RING-CH ubiquitin E3 ligase, membrane-associated RING-CH I (MARCH I), promotes the ubiquitination of the HLA-DR beta-chain. Thus, in nonactivated DCs, MARCH I induces the surface internalization of mature HLA-DR complexes, therefore reducing their stability and levels. We further demonstrate that the maturation-dependent down-regulation of MARCH I is a key event in MHC class II up-regulation at the surface of LPS-activated DCs. MARCH I is, therefore, a major regulator of HLA-DR traffic, and its loss contributes to the acquisition of the potent immunostimulatory properties of mature human DCs.


Assuntos
Células Dendríticas/imunologia , Regulação para Baixo/genética , Antígenos de Histocompatibilidade Classe II/metabolismo , Ubiquitina-Proteína Ligases/genética , Antígenos de Superfície , Transporte Biológico , Células Cultivadas , Endocitose , Antígenos HLA-DR/metabolismo , Humanos , Lipopolissacarídeos/farmacologia
13.
Cell Rep ; 36(2): 109359, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34260935

RESUMO

The anti-tumor response of Vγ9Vδ2 T cells requires the sensing of accumulated phosphoantigens (pAgs) bound intracellularly to butyrophilin 3A1 (BTN3A1). In this study, we show that butyrophilin 2A1 (BTN2A1) is required for BTN3A-mediated Vγ9Vδ2 T cell cytotoxicity against cancer cells, and that expression of the BTN2A1/BTN3A1 complex is sufficient to trigger Vγ9Vδ2 TCR activation. Also, BTN2A1 interacts with all isoforms of BTN3A (BTN3A1, BTN3A2, BTN3A3), which appears to be a rate-limiting factor to BTN2A1 export to the plasma membrane. BTN2A1/BTN3A1 interaction is enhanced by pAgs and, strikingly, B30.2 domains of both proteins are required for pAg responsiveness. BTN2A1 expression in cancer cells correlates with bisphosphonate-induced Vγ9Vδ2 T cell cytotoxicity. Vγ9Vδ2 T cell killing of cancer cells is modulated by anti-BTN2A1 monoclonal antibodies (mAbs), whose action relies on the inhibition of BTN2A1 binding to the Vγ9Vδ2TCR. This demonstrates the potential of BTN2A1 as a therapeutic target and adds to the emerging butyrophilin-family cooperation pathway in γδ T cell activation.


Assuntos
Butirofilinas/metabolismo , Proteínas de Checkpoint Imunológico/metabolismo , Neoplasias/imunologia , Neoplasias/patologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Linfócitos T Citotóxicos/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Antígenos/metabolismo , Antígenos CD/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Células HEK293 , Humanos , Ativação Linfocitária/imunologia , Camundongos , Fosforilação , Ligação Proteica , Transporte Proteico
14.
Sci Transl Med ; 13(616): eabj0835, 2021 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-34669444

RESUMO

Gamma delta T (γδ T) cells are among the most potent cytotoxic lymphocytes. Activating anti­butyrophilin 3A (BTN3A) antibodies prime diverse tumor cell types to be killed by Vγ9Vδ2 T cells, the predominant γδ T cell subset in peripheral circulation, by mechanisms independent of tumor antigen­major histocompatibility complex (MHC) complexes. In this report, we describe the development of a humanized monoclonal antibody, ICT01, with subnanomolar affinity for the three isoforms of BTN3A. We demonstrate that ICT01-activated Vγ9Vδ2 T cells kill multiple tumor cell lines and primary tumor cells, but not normal healthy cells, in an efficient process requiring approximately 20% target occupancy. We show that ICT01 activity is dependent on BTN3A and BTN2A but independent of the phosphoantigen (pAg)­binding B30.2 domain. ICT01 delays the growth of hematologic and solid tumor xenografts and prolongs survival of NOD/SCID/IL2rγnull (NSG) mice adoptively transferred with human Vγ9Vδ2 T cells. In single- and multiple-dose safety studies in cynomolgus macaques that received up to 100 mg/kg once weekly, ICT01 was well tolerated. With respect to pharmacodynamic endpoints, ICT01 selectively activated Vγ9Vδ2 T cells without affecting other BTN3A-expressing lymphocytes such as αß T or B cells. A first-in-human, phase 1/2a, open-label, clinical study of ICT01 was thus initiated in patients with advanced-stage solid tumors (EVICTION: NCT04243499; EudraCT: 2019-003847-31). Preliminary results show that ICT01 was well tolerated and pharmacodynamically active in the first patients. Digital pathology analysis of tumor biopsies of a patient with melanoma suggests that ICT01 may promote immune cell infiltration within the tumor microenvironment.


Assuntos
Ativação Linfocitária , Linfócitos T , Receptores de Antígenos de Linfócitos T gama-delta
15.
Cell Biol Int ; 33(1): 36-48, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18992834

RESUMO

Exosomes are small membrane vesicles that are released into the extracellular compartment as a consequence of fusion of multivesicular endosomes with the plasma membrane. To unravel the molecular basis of protein sorting into exosomes, we have made a chimeric protein containing the cytosolic domain of the transmembrane subunit of the viral Env protein of BLV and the ectodomain of CD8 (CDTM-BLV-CD8). When expressed in K562 cells known to constitutively secrete exosomes, the chimera was found to be very efficiently targeted to the released vesicles. Very interestingly, the cytosolic domain of the Env protein contains peptide motifs potentially recognized by components of the ESCRT machinery that could be related to chimera sorting into the vesicles. Then, quantifying the chimera secretion, we investigated the site of exosome biogenesis in K562 cells using a pharmacological approach. We present different arguments indicating that CDTM-BLV-CD8-containing exosomes are likely formed from a recycling endosomal/TGN compartment.


Assuntos
Exocitose , Exossomos/metabolismo , Produtos do Gene env/metabolismo , Vírus da Leucemia Bovina , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Bovinos , Linhagem Celular Tumoral , Membrana Celular/fisiologia , Técnica Direta de Fluorescência para Anticorpo , Produtos do Gene env/efeitos dos fármacos , Produtos do Gene env/genética , Humanos , Células K562 , Dados de Sequência Molecular , Transporte Proteico , Ratos , Receptores de Antígenos de Linfócitos T/efeitos dos fármacos , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Proteínas Recombinantes de Fusão/metabolismo , Rede trans-Golgi/metabolismo
16.
C R Biol ; 325(1): 59-65, 2002 Jan.
Artigo em Francês | MEDLINE | ID: mdl-11862623

RESUMO

Cell cultures represent versatile and useful experimental models of transmissible spongiform encephalopathies. These models include chronically prion infected cell lines, as well as cultures expressing variable amounts of wild-type, mutated or chimeric prion proteins. These cultures have been widely used to investigate the biology of both the normal and the pathological isoform of the prion protein. They have also contributed to the comprehension of the pathogenic processes occurring in transmissible spongiform encephalopathies and in the development of new therapeutic approaches of these diseases.


Assuntos
Técnicas de Cultura de Células , Modelos Biológicos , Doenças Priônicas , Animais , Linhagem Celular , Expressão Gênica , Camundongos , Camundongos Transgênicos , Mutação , Príons/genética , Transfecção
17.
Biocell ; 26(2): 205-15, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12240554

RESUMO

During the differentiation of erythroid cells, a vast program of maturation takes place, leading to decay or elimination of organelles, including the nucleus, mitochondria, ribosomes, lysosomes, endoplasmic reticulum and Golgi apparatus. During the last step of red cell maturation, remaining organelles, primarily mitochondria and ribosomes but also vestiges of others are finally cleared from the cell. This cleaning session also affects specific proteins that are partially or entirely removed from the cell surface. The interplay of the various events and their causal relationships are approached here.


Assuntos
Mitocôndrias/metabolismo , Reticulócitos/metabolismo , Trifosfato de Adenosina/metabolismo , Araquidonato 15-Lipoxigenase/metabolismo , Caspases/metabolismo , Membrana Celular/metabolismo , Ativação Enzimática , Complexo de Golgi/metabolismo , Modelos Biológicos , Proteínas/metabolismo , Espécies Reativas de Oxigênio , Receptores da Transferrina/metabolismo , Relação Estrutura-Atividade , Temperatura
18.
Methods Mol Biol ; 960: 517-527, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23329511

RESUMO

Ubiquitinated protein detection is often troublesome since in most cases this modification reduces the half-life of targeted proteins, inducing their degradation. Furthermore, ubiquitination is reversible thanks to the action of highly specific deubiquitinases present in all eukaryotic cells. MHC molecules ubiquitination has been demonstrated to be a key event in the regulation of the potent immunostimulatory properties of activated human dendritic cells.


Assuntos
Antígenos de Histocompatibilidade Classe II/metabolismo , Ubiquitinação , Western Blotting , Células Dendríticas/metabolismo , Eletroforese em Gel de Poliacrilamida , Células HeLa , Antígenos de Histocompatibilidade Classe II/isolamento & purificação , Humanos , Imunoprecipitação , Monócitos/citologia
20.
Autophagy ; 8(3): 350-63, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22377621

RESUMO

A significant portion of newly synthesized protein fails to fold properly and is quickly degraded. These defective ribosomal products (DRiPs) are substrates for the ubiquitin-proteasome system (UPS) and give rise to a large fraction of peptides presented by major histocompatibility complex class I molecules (MHCI). Here, we showed that DRiPs are also autophagy substrates, which accumulate upon autophagy inhibition in aggresome-like-induced structures (ALIS). Aggregation is critically depending on p62/SQSTM1, but occurs in the absence of activation of the NRF2 signaling axis and transcriptional regulation of p62/SQSTM1. We demonstrated that autophagy-targeted DRiPs can become UPS substrates and give rise to MHCI presented peptides upon autophagy inhibition. We further demonstrated that autophagy targeting of DRiPs is controlled by NBR1, but not p62/SQSTM1, CHIP or BAG-1. Active autophagy therefore directly modulates MHCI presentation by constantly degrading endogenous defective neosynthesized antigens, which are submitted to at least two distinct quality control mechanisms.


Assuntos
Apresentação de Antígeno/imunologia , Autofagia , Antígenos de Histocompatibilidade Classe I/imunologia , Corpos de Inclusão/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Biossíntese de Proteínas , Processamento de Proteína Pós-Traducional , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apresentação de Antígeno/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Corpos de Inclusão/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular , Chaperonas Moleculares/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Inibidores de Proteassoma/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Dobramento de Proteína/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Estrutura Quaternária de Proteína , Proteínas/metabolismo , Proteólise/efeitos dos fármacos , Puromicina/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Ribossomos/metabolismo , Proteína Sequestossoma-1 , Transdução de Sinais/efeitos dos fármacos , Especificidade por Substrato/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA