Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.883
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 49(6): 1132-1147.e7, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30552022

RESUMO

Serrated adenocarcinoma, an alternative pathway for colorectal cancer (CRC) development, accounts for 15%-30% of all CRCs and is aggressive and treatment resistant. We show that the expression of atypical protein kinase C ζ (PKCζ) and PKCλ/ι was reduced in human serrated tumors. Simultaneous inactivation of the encoding genes in the mouse intestinal epithelium resulted in spontaneous serrated tumorigenesis that progressed to advanced cancer with a strongly reactive and immunosuppressive stroma. Whereas epithelial PKCλ/ι deficiency led to immunogenic cell death and the infiltration of CD8+ T cells, which repressed tumor initiation, PKCζ loss impaired interferon and CD8+ T cell responses, which resulted in tumorigenesis. Combined treatment with a TGF-ß receptor inhibitor plus anti-PD-L1 checkpoint blockade showed synergistic curative activity. Analysis of human samples supported the relevance of these kinases in the immunosurveillance defects of human serrated CRC. These findings provide insight into avenues for the detection and treatment of this poor-prognosis subtype of CRC.


Assuntos
Mucosa Intestinal/imunologia , Neoplasias Intestinais/imunologia , Isoenzimas/imunologia , Proteína Quinase C/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/imunologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/metabolismo , Feminino , Humanos , Vigilância Imunológica/genética , Vigilância Imunológica/imunologia , Mucosa Intestinal/enzimologia , Mucosa Intestinal/patologia , Neoplasias Intestinais/enzimologia , Neoplasias Intestinais/genética , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos Knockout , Camundongos Transgênicos , Pessoa de Meia-Idade , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo
2.
Genes Dev ; 31(11): 1109-1121, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28698296

RESUMO

A key feature of high-grade serous ovarian carcinoma (HGSOC) is frequent amplification of the 3q26 locus harboring PRKC-ι (PRKCI). Here, we show that PRKCI is also expressed in early fallopian tube lesions, called serous tubal intraepithelial carcinoma. Transgenic mouse studies establish PRKCI as an ovarian cancer-specific oncogene. Mechanistically, we show that the oncogenic activity of PRKCI relates in part to the up-regulation of TNFα to promote an immune-suppressive tumor microenvironment characterized by an abundance of myeloid-derived suppressor cells and inhibition of cytotoxic T-cell infiltration. Furthermore, system-level and functional analyses identify YAP1 as a downstream effector in tumor progression. In human ovarian cancers, high PRKCI expression also correlates with high expression of TNFα and YAP1 and low infiltration of cytotoxic T cells. The PRKCI-YAP1 regulation of the tumor immunity provides a therapeutic strategy for highly lethal ovarian cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , Tolerância Imunológica/genética , Isoenzimas/genética , Isoenzimas/imunologia , Neoplasias Ovarianas/genética , Proteína Quinase C/genética , Proteína Quinase C/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas de Ciclo Celular , Movimento Celular/genética , Citocinas/genética , Feminino , Humanos , Isoenzimas/metabolismo , Camundongos , Camundongos Transgênicos , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/fisiopatologia , Fosfoproteínas/metabolismo , Proteína Quinase C/metabolismo , Linfócitos T Citotóxicos/citologia , Linfócitos T Citotóxicos/imunologia , Microambiente Tumoral/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Proteínas de Sinalização YAP
3.
Nat Immunol ; 13(11): 1045-1054, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23023391

RESUMO

Lipopolysaccharide activates plasma-membrane signaling and endosomal signaling by Toll-like receptor 4 (TLR4) through the TIRAP-MyD88 and TRAM-TRIF adaptor complexes, respectively, but it is unclear how the signaling switch between these cell compartments is coordinated. In dendritic cells, we found that the p110δ isoform of phosphatidylinositol-3-OH kinase (PI(3)K) induced internalization of TLR4 and dissociation of TIRAP from the plasma membrane, followed by calpain-mediated degradation of TIRAP. Accordingly, inactivation of p110δ prolonged TIRAP-mediated signaling from the plasma membrane, which augmented proinflammatory cytokine production while decreasing TRAM-dependent endosomal signaling that generated anti-inflammatory cytokines (interleukin 10 and interferon-ß). In line with that altered signaling output, p110δ-deficient mice showed enhanced endotoxin-induced death. Thus, by controlling the 'topology' of TLR4 signaling complexes, p110δ balances overall homeostasis in the TLR4 pathway.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/imunologia , Células Dendríticas/imunologia , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Calpaína/farmacologia , Compartimento Celular/imunologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Membrana Celular/imunologia , Células Cultivadas , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Endossomos/genética , Endossomos/imunologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Interferon beta/biossíntese , Interferon beta/imunologia , Interleucina-10/biossíntese , Interleucina-10/imunologia , Isoenzimas/genética , Isoenzimas/imunologia , Lipopolissacarídeos/farmacologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Receptores de Interleucina/genética , Receptores de Interleucina/imunologia , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/imunologia , Choque Séptico/genética , Choque Séptico/imunologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Receptor 4 Toll-Like/genética
4.
Nat Immunol ; 12(7): 647-54, 2011 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-21602810

RESUMO

Polarization of the T cell microtubule-organizing center (MTOC) toward the antigen-presenting cell (APC) is driven by the accumulation of diacylglycerol (DAG) at the immunological synapse (IS). The mechanisms that couple DAG to the MTOC are not known. By single-cell photoactivation of the T cell antigen receptor (TCR), we found that three distinct isoforms of protein kinase C (PKC) were recruited by DAG to the IS in two steps. PKC-ɛ and PKC-η accumulated first in a broad region of membrane, whereas PKC-θ arrived later in a smaller zone. Functional experiments indicated that PKC-θ was required for MTOC reorientation and that PKC-ɛ and PKC-η operated redundantly to promote the recruitment of PKC-θ and subsequent polarization responses. Our results establish a previously uncharacterized role for PKC proteins in T cell polarity.


Assuntos
Polaridade Celular/imunologia , Citoesqueleto/enzimologia , Isoenzimas/imunologia , Proteína Quinase C-épsilon/imunologia , Proteína Quinase C/imunologia , Linfócitos T/enzimologia , Animais , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/enzimologia , Células Apresentadoras de Antígenos/imunologia , Citoesqueleto/imunologia , Diglicerídeos/imunologia , Sinapses Imunológicas/enzimologia , Sinapses Imunológicas/imunologia , Camundongos , Camundongos Transgênicos , Proteína Quinase C-theta , Receptores de Antígenos de Linfócitos T/imunologia , Análise de Célula Única , Linfócitos T/citologia , Linfócitos T/imunologia
5.
Nat Immunol ; 12(11): 1105-12, 2011 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-21964608

RESUMO

Protein kinase C-θ (PKC-θ) translocates to the center of the immunological synapse, but the underlying mechanism and its importance in T cell activation are unknown. Here we found that the V3 domain of PKC-θ was necessary and sufficient for localization to the immunological synapse mediated by association with the coreceptor CD28 and dependent on the kinase Lck. We identified a conserved proline-rich motif in V3 required for association with CD28 and immunological synapse localization. We found association with CD28 to be essential for PKC-θ-mediated downstream signaling and the differentiation of T helper type 2 cells (T(H)2 cells) and interleukin 17-producing helper T cells (T(H)17 cells) but not of T helper type 1 cells (T(H)1 cells). Ectopic expression of V3 sequestered PKC-θ from the immunological synapse and interfered with its functions. Our results identify a unique mode of CD28 signaling, establish a molecular basis for the immunological synapse localization of PKC-θ and indicate V3-based 'decoys' may be therapeutic modalities for T cell-mediated inflammatory diseases.


Assuntos
Antígenos CD28/metabolismo , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Subpopulações de Linfócitos T/metabolismo , Células Th17/metabolismo , Células Th2/metabolismo , Motivos de Aminoácidos/genética , Animais , Antígenos CD28/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Sinapses Imunológicas , Imunomodulação , Isoenzimas/genética , Isoenzimas/imunologia , Ativação Linfocitária , Camundongos , Camundongos Knockout , Domínios Proteicos Ricos em Prolina/genética , Ligação Proteica/imunologia , Proteína Quinase C/genética , Proteína Quinase C/imunologia , Proteína Quinase C-theta , Transporte Proteico/imunologia , Transdução de Sinais/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/patologia , Células Th17/imunologia , Células Th17/patologia , Células Th2/imunologia , Células Th2/patologia
6.
Nat Immunol ; 12(11): 1113-8, 2011 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-21983831

RESUMO

Protein kinase C-θ (PKC-θ) is required for activation of the transcription factor NF-κB induced by signaling via the T cell antigen receptor (TCR); however, the direct activator of PKC-θ is unknown. We report that the kinase GLK (MAP4K3) directly activated PKC-θ during TCR signaling. TCR signaling activated GLK by inducing its direct interaction with the upstream adaptor SLP-76. GLK-deficient mice had impaired immune responses and were resistant to experimental autoimmune encephalomyelitis. Consistent with that, people with systemic lupus erythematosus had considerable enhanced GLK expression and activation of PKC-θ and the kinase IKK in T cells, and the frequency of GLK-overexpressing T cells was directly correlated with disease severity. Thus, GLK is a direct activator of PKC-θ, and activation of GLK-PKC-θ-IKK could be used as new diagnostic biomarkers and therapeutic targets for systemic lupus erythematosus.


Assuntos
Isoenzimas/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , NF-kappa B/metabolismo , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Linfócitos T/metabolismo , Adulto , Animais , Autoimunidade/genética , Progressão da Doença , Feminino , Humanos , Isoenzimas/genética , Isoenzimas/imunologia , Células Jurkat , Ativação Linfocitária/genética , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , NF-kappa B/imunologia , Proteína Quinase C/genética , Proteína Quinase C/imunologia , Proteína Quinase C-theta , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , RNA Interferente Pequeno/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia
7.
J Immunol ; 202(10): 2971-2981, 2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-30944160

RESUMO

Phagocytosis is an essential element of the immune response, assuring the elimination of pathogens, cellular debris, and apoptotic and tumoral cells. Activation of phagocytosis by the FcγR stimulates phospholipase D (PLD) activity and triggers the production of phosphatidic acid (PA) at the plasma membrane of macrophages, but the regulatory mechanisms involved are still not clearly understood. In this study, we examined the role of the small GTPase Arf6 in the activation of the PLD isoforms during FcγR-mediated phagocytosis. In RAW 264.7 macrophage cells, expressed Arf6-GFP partially colocalized with PLD1-hemagglutinin on intracellular membrane-bound vesicles and with PLD2-hemagglutinin at the plasma membrane. Both PLD isoforms were found to interact with Arf6 during FcγR-mediated phagocytosis as seen by immunoprecipitation experiments. In macrophages stimulated for phagocytosis, Arf6 was observed to be associated with nascent phagosomes. RNA interference knockdown of Arf6 reduced the amount of active Arf6 associated with phagosomes, revealed by the MT2-GFP probe that specifically binds to Arf6-GTP. Arf6 silencing concomitantly decreased PLD activity as well as the levels of PA found on phagosomes and phagocytic sites as shown with the PA probe Spo20p-GFP. Altogether, our results indicate that Arf6 is involved in the regulation of PLD activity and PA synthesis required for efficient phagocytosis.


Assuntos
Fatores de Ribosilação do ADP/imunologia , Macrófagos/imunologia , Fagocitose , Fosfolipase D/imunologia , Receptores de IgG/imunologia , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/genética , Animais , Isoenzimas/genética , Isoenzimas/imunologia , Macrófagos/citologia , Camundongos , Fagossomos/genética , Fagossomos/imunologia , Ácidos Fosfatídicos/genética , Ácidos Fosfatídicos/imunologia , Fosfolipase D/genética , Células RAW 264.7 , Receptores de IgG/genética
8.
Nucleic Acids Res ; 47(16): 8632-8648, 2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31392984

RESUMO

CRISPR-Cas systems provide heritable immunity against viruses by capturing short invader DNA sequences, termed spacers, and incorporating them into the CRISPR loci of the prokaryotic host genome. Here, we investigate DNA elements that control accurate spacer uptake in the type II-A CRISPR locus of Streptococcus thermophilus. We determined that purified Cas1 and Cas2 proteins catalyze spacer integration with high specificity for CRISPR repeat junctions. We show that 10 bp of the CRISPR leader sequence is critical for stimulating polarized integration preferentially at the repeat proximal to the leader. Spacer integration proceeds through a two-step transesterification reaction where the 3' hydroxyl groups of the spacer target both repeat borders on opposite strands. The leader-proximal end of the repeat is preferentially targeted for the first site of integration through recognition of sequences spanning the leader-repeat junction. Subsequently, second-site integration at the leader-distal end of the repeat is specified by multiple determinants including a length-defining mechanism relying on a repeat element proximal to the second site of integration. Our results highlight the intrinsic ability of type II Cas1/Cas2 proteins to coordinate directional and site-specific spacer integration into the CRISPR locus to ensure precise duplication of the repeat required for CRISPR immunity.


Assuntos
Sistemas CRISPR-Cas , Endonucleases/genética , Edição de Genes , Genoma Bacteriano , Streptococcus thermophilus/genética , Sequência de Bases , Endonucleases/imunologia , Endonucleases/metabolismo , Esterificação , Loci Gênicos , Isoenzimas/genética , Isoenzimas/imunologia , Isoenzimas/metabolismo , Mutagênese Insercional , Plasmídeos/química , Plasmídeos/metabolismo , Streptococcus thermophilus/imunologia , Streptococcus thermophilus/metabolismo , Streptococcus thermophilus/virologia , Vírus/genética , Vírus/metabolismo
9.
Cancer Immunol Immunother ; 69(3): 449-463, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31932876

RESUMO

Lactate dehydrogenase C (LDHC) is an archetypical cancer testis antigen with limited expression in adult tissues and re-expression in tumors. This restricted expression pattern together with the important role of LDHC in cancer metabolism renders LDHC a potential target for immunotherapy. This study is the first to investigate the immunogenicity of LDHC using T cells from healthy individuals. LDHC-specific T cell responses were induced by in vitro stimulation with synthetic peptides, or by priming with autologous peptide-pulsed dendritic cells. We evaluated T cell activation by IFN-γ ELISpot and determined cytolytic activity of HLA-A*0201-restricted T cells in breast cancer cell co-cultures. In vitro T cell stimulation induced IFN-γ secretion in response to numerous LDHC-derived peptides. Analysis of HLA-A*0201 responses revealed a significant T cell activation after stimulation with peptide pools 2 (PP2) and 8 (PP8). The PP2- and PP8-specific T cells displayed cytolytic activity against breast cancer cells with endogenous LDHC expression within a HLA-A*0201 context. We identified peptides LDHC41-55 and LDHC288-303 from PP2 and PP8 to elicit a functional cellular immune response. More specifically, we found an increase in IFN-γ secretion by CD8 + T cells and cancer-cell-killing of HLA-A*0201/LDHC positive breast cancer cells by LDHC41-55- and LDHC288-303-induced T cells, albeit with a possible antigen recognition threshold. The majority of induced T cells displayed an effector memory phenotype. To conclude, our findings support the rationale to assess LDHC as a targetable cancer testis antigen for immunotherapy, and in particular the HLA-A*0201 restricted LDHC41-55 and LDHC288-303 peptides within LDHC.


Assuntos
Epitopos de Linfócito T/imunologia , Antígeno HLA-A2/imunologia , Imunoterapia/métodos , L-Lactato Desidrogenase/imunologia , Linhagem Celular Tumoral , Feminino , Humanos , Isoenzimas/imunologia , Masculino
10.
FASEB J ; 33(11): 12780-12799, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31480861

RESUMO

Intercellular communication between lymphocytes plays a fundamental role in numerous immune responses. Previously, we demonstrated that hyperhomocysteinemia (HHcy) induced T cell intracellular glycolytic-lipogenic reprogramming and IFN-γ secretion via pyruvate kinase muscle isozyme 2 (PKM2) to accelerate atherosclerosis. Usually, B cells partially obtain help from T cells in antibody responses. However, whether PKM2 activation in T cells regulates B cell antibody production is unknown. Extracellular vesicles (EVs) are important cellular communication vehicles. Here, we found that PKM2 activator TEPP46-stimulated T-cell-derived EVs promoted B-cell IgG secretion. Conversely, EVs secreted from PKM2-null T cells were internalized into B cells and markedly inhibited B-cell mitochondrial programming, activation, and IgG production. Mechanistically, lipidomics analyses showed that increased ceramides in PKM2-activated T-cell EVs were mainly responsible for enhanced B cell IgG secretion induced by these EVs. Finally, quantum dots (QDs) were packaged with PKM2-null T cell EVs and anti-CD19 antibody to exert B-cell targeting and inhibit IgG production, eventually ameliorating HHcy-accelerated atherosclerosis in vivo. Thus, PKM2-mediated EV ceramides in T cells may be an important cargo for T-cell-regulated B cell IgG production, and QD-CD19-PKM2-null T cell EVs hold high potential to treat B cell overactivation-related diseases.-Yang, J., Dang, G., Lü, S., Liu, H., Ma, X., Han, L., Deng, J., Miao, Y., Li, X., Shao, F., Jiang, C., Xu, Q., Wang, X., Feng, J. T-cell-derived extracellular vesicles regulate B-cell IgG production via pyruvate kinase muscle isozyme 2.


Assuntos
Formação de Anticorpos , Linfócitos B/imunologia , Vesículas Extracelulares/imunologia , Imunoglobulina G/imunologia , Piruvato Quinase/imunologia , Linfócitos T/imunologia , Animais , Linfócitos B/patologia , Vesículas Extracelulares/patologia , Feminino , Doenças do Sistema Imunitário/imunologia , Doenças do Sistema Imunitário/patologia , Doenças do Sistema Imunitário/terapia , Isoenzimas/imunologia , Camundongos , Camundongos Knockout para ApoE , Pontos Quânticos , Linfócitos T/patologia
11.
FASEB J ; 33(5): 6539-6550, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30807229

RESUMO

As a master regulator for metabolic and energy homeostasis, AMPK controls the activity of metabolic enzymes and transcription factors in response to cellular ATP status. AMPK has been thus recognized as a main target for the regulation of cellular energy metabolism. Here, we report that AMPK can be down-regulated by the cullin-RING ubiquitin E3 ligase 4A (CRL4A) with cereblon (CRBN). CRL4A interacted with AMPK holoenzymes and mediated AMPKα-specific polyubiquitination for its proteasomal degradation through non-K48 polyubiquitin linkages. In the ubiquitination system, CRBN was required for efficient polyubiquitination of AMPKα subunits. Consistently, polyubiquitination of AMPKα subunits was reduced by inhibitors of CRL4A-CRBN. Physiologic function of AMPK down-regulation by CRL4-CRBN was also confirmed using mouse bone marrow-derived mast cells (BMMCs). The inactivation of CRL4A-CRBN in BMMC increased AMPK stability and suppressed secretion of allergic mediators via AMPK activation followed by MAPK inhibition. In addition, CRBN knockout of BMMC also decreased allergic responses in mice. Our results suggest that the CRL4A-CRBN axis could be a target for the regulation of AMPK-dependent responses.-Kwon, E., Li, X., Deng, Y., Chang, H. W., Kim, D. Y. AMPK is down-regulated by the CRL4A-CRBN axis through the polyubiquitination of AMPKα isoforms.


Assuntos
Proteínas Quinases Ativadas por AMP/imunologia , Células da Medula Óssea/imunologia , Regulação para Baixo/imunologia , Mastócitos/imunologia , Transdução de Sinais/imunologia , Complexos Ubiquitina-Proteína Ligase/imunologia , Ubiquitinação/imunologia , Proteínas Quinases Ativadas por AMP/genética , Animais , Células da Medula Óssea/patologia , Células HEK293 , Humanos , Hipersensibilidade/genética , Hipersensibilidade/imunologia , Hipersensibilidade/patologia , Isoenzimas/genética , Isoenzimas/imunologia , Mastócitos/patologia , Camundongos , Camundongos Knockout , Transdução de Sinais/genética , Complexos Ubiquitina-Proteína Ligase/genética , Ubiquitinação/genética
12.
EMBO Rep ; 19(1): 89-101, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29191979

RESUMO

Shigella deploys a unique mechanism to manipulate macrophage pyroptosis by delivering the IpaH7.8 E3 ubiquitin ligase via its type III secretion system. IpaH7.8 ubiquitinates glomulin (GLMN) and elicits its degradation, thereby inducing inflammasome activation and pyroptotic cell death of macrophages. Here, we show that GLMN specifically binds cellular inhibitor of apoptosis proteins 1 and 2 (cIAP1 and cIAP2), members of the inhibitor of apoptosis (IAP) family of RING-E3 ligases, which results in reduced E3 ligase activity, and consequently inflammasome-mediated death of macrophages. Importantly, reducing the levels of GLMN in macrophages via IpaH7.8, or siRNA-mediated knockdown, enhances inflammasome activation in response to infection by Shigella, Salmonella, or Pseudomonas, stimulation with NLRP3 inflammasome activators (including SiO2, alum, or MSU), or stimulation of the AIM2 inflammasome by poly dA:dT GLMN binds specifically to the RING domain of both cIAPs, which inhibits their self-ubiquitination activity. These findings suggest that GLMN is a negative regulator of cIAP-mediated inflammasome activation, and highlight a unique Shigella stratagem to kill macrophages, promoting severe inflammation.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Interações Hospedeiro-Patógeno , Inflamassomos/genética , Proteínas Inibidoras de Apoptose/genética , Macrófagos/microbiologia , Proteínas Musculares/genética , Shigella flexneri/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Regulação da Expressão Gênica , Inflamassomos/imunologia , Proteínas Inibidoras de Apoptose/imunologia , Isoenzimas/genética , Isoenzimas/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Musculares/imunologia , Cultura Primária de Células , Ligação Proteica , Piroptose/genética , Salmonella typhimurium/crescimento & desenvolvimento , Salmonella typhimurium/imunologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Shigella flexneri/crescimento & desenvolvimento , Transdução de Sinais , Sistemas de Secreção Tipo III/genética , Sistemas de Secreção Tipo III/imunologia
13.
Proc Natl Acad Sci U S A ; 114(49): E10560-E10567, 2017 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-29158395

RESUMO

In B cells, Ig class switch recombination (CSR) is initiated by activation-induced cytidine deaminase (AID), the activity of which leads to DNA double-strand breaks (DSBs) within IgH switch (S) regions. Preferential targeting of AID-mediated DSBs to S sequences is critical for allowing diversification of antibody functions, while minimizing potential off-target oncogenic events. Here, we used gene targeted inactivation of histone methyltransferase (HMT) multiple myeloma SET domain (MMSET) in mouse B cells and the CH12F3 cell line to explore its role in CSR. We find that deletion of MMSET-II, the isoform containing the catalytic SET domain, inhibits CSR without affecting either IgH germline transcription or joining of DSBs within S regions by classical nonhomologous end joining (C-NHEJ). Instead, we find that MMSET-II inactivation leads to decreased AID recruitment and DSBs at the upstream donor Sµ region. Our findings suggest a role for the HMT MMSET in promoting AID-mediated DNA breaks during CSR.


Assuntos
Citidina Desaminase/genética , DNA/genética , Histona-Lisina N-Metiltransferase/genética , Switching de Imunoglobulina , Região de Troca de Imunoglobulinas , Imunoglobulinas/genética , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Domínio Catalítico , Citidina Desaminase/imunologia , DNA/imunologia , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Regulação da Expressão Gênica , Inativação Gênica , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/imunologia , Imunoglobulinas/metabolismo , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/imunologia , Camundongos , Camundongos Knockout , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Recombinação Genética , Transdução de Sinais
14.
Proc Natl Acad Sci U S A ; 114(48): 12791-12796, 2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29133416

RESUMO

Metastasis is a major cause of cancer-related deaths. Approximately 80% of patients with colorectal cancer develop liver metastasis and 20% develop lung metastasis. We found that at different stages of colon cancer, IFNγ secretion from peripheral blood mononuclear cells was decreased compared with healthy controls. The ribosomal S6 kinase (RSK) family of kinases has multiple cellular functions, and we examined their roles in this observed IFNγ decrease. Flow cytometry analysis of wild-type (WT) and RSK2 knockout (KO) mice revealed significantly lower levels of IFNγ in the RSK2 KO mice compared with the WT mice. Since IFNγ is a component of immunity, which contributes to protection against metastatic carcinomas, we conducted a colon cancer liver metastasis experiment. We found significantly greater metastasis in RSK2 KO mice compared with WT mice. Transcription factor T-bet can directly activate Ifnγ gene transcription. In vitro kinase assay results showed that RSK2 phosphorylated T-bet at serines 498 and 502. We show that phosphorylation of T-bet by RSK2 is required for IFNγ expression, because knockdown of RSK2 expression or overexpression of mutant T-bet reduces IFNγ mRNA expression. To verify the function of the phosphorylation sites, we overexpressed a constitutively active mutant T-bet (S498E/S502E) in bone marrow. Mutant T-bet restored the IFNγ mRNA levels and dramatically reduced the metastasis rate in these mice. Overall, these results indicate that phosphorylation of T-bet is required for the inhibition of colon cancer metastasis and growth through a positive regulation of RSK2/T-bet/IFNγ signaling.


Assuntos
Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica , Interferon gama/genética , Neoplasias Hepáticas/genética , Neoplasias Pulmonares/genética , Proteínas Quinases S6 Ribossômicas/genética , Proteínas com Domínio T/genética , Animais , Transplante de Medula Óssea , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Feminino , Humanos , Interferon gama/imunologia , Isoenzimas/genética , Isoenzimas/imunologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/patologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/prevenção & controle , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Fosforilação , Proteínas Quinases S6 Ribossômicas/imunologia , Serina/metabolismo , Transdução de Sinais , Proteínas com Domínio T/imunologia , Transfecção , Irradiação Corporal Total
15.
Int J Mol Sci ; 21(16)2020 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-32806627

RESUMO

Fabry Disease (FD) is a rare, X-linked, lysosomal storage disease that mainly causes renal, cardiac and cerebral complications. Enzyme replacement therapy (ERT) with recombinant alpha-galactosidase A is available, but approximately 50% of male patients with classical FD develop inhibiting anti-drug antibodies (iADAs) that lead to reduced biochemical responses and an accelerated loss of renal function. Once immunization has occurred, iADAs tend to persist and tolerization is hard to achieve. Here we developed a pre-treatment prediction model for iADA development in FD using existing data from 120 classical male FD patients from three European centers, treated with ERT. We found that nonsense and frameshift mutations in the α-galactosidase A gene (p = 0.05), higher plasma lysoGb3 at baseline (p < 0.001) and agalsidase beta as first treatment (p = 0.006) were significantly associated with iADA development. Prediction performance of a Random Forest model, using multiple variables (AUC-ROC: 0.77) was compared to a logistic regression (LR) model using the three significantly associated variables (AUC-ROC: 0.77). The LR model can be used to determine iADA risk in individual FD patients prior to treatment initiation. This helps to determine in which patients adjusted treatment and/or immunomodulatory regimes may be considered to minimize iADA development risk.


Assuntos
Anticorpos/imunologia , Doença de Fabry/tratamento farmacológico , Doença de Fabry/imunologia , Isoenzimas/imunologia , Isoenzimas/uso terapêutico , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico , alfa-Galactosidase/imunologia , alfa-Galactosidase/uso terapêutico , Adolescente , Adulto , Algoritmos , Área Sob a Curva , Criança , Estudos de Coortes , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Curva ROC , Fatores de Risco , Adulto Jovem
16.
Int J Mol Sci ; 21(19)2020 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-33003611

RESUMO

Fabry disease (FD) is an X-linked lysosomal disorder caused by mutations in GLA gene resulting in lack of or faulty α-galactosidase A (α-GalA) enzyme. Enzyme replacement therapy (ERT) with recombinant human α-GalA enzyme (agalsidase) is the standard treatment option for FD. Infusion-related reactions (IRRs), with symptoms ranging from rigors, to fever, pain, vomiting, angioedema and diarrhea, are often seen due to immune response against the exogenous enzyme. To elucidate the mechanisms causing the IRRs in FD, eight patients who developed IRRs were investigated. All, except one, tested negative for agalsidase-specific IgE and had normal tryptase levels. Circulating dendritic cells were drastically reduced during IRRs, suggesting possible sequestration to the sites of inflammation. An increase in NK cells and a decrease in T cells were also observed. Cytokines IL-4, IL-8 and TNF-α showed a significant increase, indicating nonspecific degranulation of mast cells. All IRRs were managed successfully using a combination of standard premedications and mast cell stabilizers without any interruption of therapy. Taken together, the results indicate crosstalk between immune cells resulting in IgE-independent mast-cell-specific allergic inflammation. Mast cell stabilizers could be used to control IRRs and for safe reintroduction of agalsidase in patients previously treated with ERT.


Assuntos
Doença de Fabry/tratamento farmacológico , Doença de Fabry/imunologia , Inflamação/imunologia , Isoenzimas/imunologia , Proteínas Recombinantes/imunologia , alfa-Galactosidase/imunologia , Imunidade Adaptativa/efeitos dos fármacos , Imunidade Adaptativa/imunologia , Adolescente , Adulto , Criança , Terapia de Reposição de Enzimas/efeitos adversos , Doença de Fabry/genética , Doença de Fabry/patologia , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Imunoglobulina E/imunologia , Inflamação/patologia , Reação no Local da Injeção/genética , Reação no Local da Injeção/imunologia , Isoenzimas/administração & dosagem , Masculino , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , Pessoa de Meia-Idade , Proteínas Recombinantes/administração & dosagem , Adulto Jovem , alfa-Galactosidase/administração & dosagem
17.
Cell Immunol ; 340: 103915, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31054776

RESUMO

During viral infection, the innate immune system represents the first defense line of the human body. The pathogen associated molecular patterns (PAMPs) from the viruses are recognized by pattern recognition receptors (PRRs) of the host cell, especially from those of the immune cells. Sensing of PAMPs by PRRs elicits an elegant signal transduction system, ultimately leading to the production of type I interferons (IFNs) and proinflammatory cytokines. Ubiquitination, with its versatile functions, plays a central role in modulating almost every single step of this signaling cascade. Ubiquitin ligases, which catalyze different types of ubiquitination correlating with multiple functions, are the key participant in fine-tuning antiviral signal transduction. In this review, we focus on summarizing the ubiquitin ligases that regulate the key signaling molecules in antiviral innate immunity.


Assuntos
Imunidade Inata , Isoenzimas/genética , Processamento de Proteína Pós-Traducional , Receptores de Reconhecimento de Padrão/genética , Ubiquitina-Proteína Ligases/genética , Viroses/imunologia , Animais , Citocinas/genética , Citocinas/imunologia , Humanos , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Isoenzimas/imunologia , Linfócitos/enzimologia , Linfócitos/imunologia , Linfócitos/virologia , Camundongos , NF-kappa B/genética , NF-kappa B/imunologia , Moléculas com Motivos Associados a Patógenos/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores de Reconhecimento de Padrão/imunologia , Transdução de Sinais , Ubiquitina-Proteína Ligases/imunologia , Ubiquitinação , Viroses/enzimologia , Viroses/genética , Viroses/virologia
18.
Cell Immunol ; 340: 103922, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31078284

RESUMO

T cells play critical roles in immune responses to pathogens, autoimmunity, and antitumor immunity. During the past few decades, increasing numbers of studies have demonstrated the significance of protein ubiquitination in T cell-mediated immunity. Several E3 ubiquitin ligases and deubiquitinases (DUBs) have been identified as either positive or negative regulators of T cell development and function. In this review, we mainly focus on the roles of DUBs (especially ubiquitin-specific proteases (USPs)) in modulating T cell differentiation and function, as well as the molecular mechanisms. Understanding how T cell development and function is regulated by ubiquitination and deubiquitination will provide novel strategies for treating infection, autoimmune diseases, and cancer.


Assuntos
Doenças Autoimunes/imunologia , Enzimas Desubiquitinantes/genética , Neoplasias/imunologia , Processamento de Proteína Pós-Traducional , Linfócitos T/imunologia , Ubiquitina-Proteína Ligases/genética , Animais , Doenças Autoimunes/enzimologia , Doenças Autoimunes/genética , Doenças Autoimunes/patologia , Diferenciação Celular , Proliferação de Células , Enzimas Desubiquitinantes/imunologia , Humanos , Tolerância Imunológica , Imunidade Celular , Isoenzimas/genética , Isoenzimas/imunologia , Camundongos , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Transdução de Sinais , Linfócitos T/enzimologia , Linfócitos T/patologia , Ubiquitina-Proteína Ligases/imunologia , Ubiquitinação
19.
Cell Immunol ; 340: 103905, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30827673

RESUMO

Ubiquitylation is a post-translational modification (PTM) that controls various cellular signaling pathways. It is orchestrated by a three-step enzymatic cascade know as the ubiquitin proteasome system (UPS). E3 ligases dictate the specificity to the substrates, primarily leading to proteasome-dependent degradation. Deregulation of the UPS components by various mechanisms contributes to the pathogenesis of cancer. This review focuses on E3 ligase-substrates pairings that are implicated in B-cell malignancies. Understanding the molecular mechanism of specific E3 ubiquitin ligases will present potential opportunities for the development of targeted therapeutic approaches.


Assuntos
Leucemia de Células B/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Processamento de Proteína Pós-Traducional , Ubiquitina-Proteína Ligases/genética , Antineoplásicos Imunológicos/uso terapêutico , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/patologia , Bortezomib/uso terapêutico , Humanos , Isoenzimas/genética , Isoenzimas/imunologia , Leucemia de Células B/tratamento farmacológico , Leucemia de Células B/genética , Leucemia de Células B/patologia , Terapia de Alvo Molecular/métodos , NF-kappa B/genética , NF-kappa B/imunologia , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais , Ubiquitina-Proteína Ligases/imunologia , Ubiquitinação
20.
Mol Genet Metab ; 126(2): 162-168, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30473480

RESUMO

BACKGROUND: Treatment of Fabry disease (FD) with recombinant alpha-galactosidase A (r-αGAL A) is complicated by the formation of anti-drug antibodies in the majority of male patients with the classical disease phenotype. Detailed information regarding antibody subtypes, onset and persistence of antibody development and their effect on treatment efficacy is sparse. METHODS: A retrospective study was carried out in 39 male patients with classical FD, treated with either agalsidase-alfa or agalsidase-beta (mean follow up of 10 years). With six to twelve months intervals plasma-induced in vitro inhibition of enzyme activity, lysoglobotriaosylsphingosine (lysoGb3) levels and renal function were assessed. In a subset of 12 patients, additionally anti- r-αGAL A IgM, IgA and IgG1, 2, 3 and 4 levels were analyzed. RESULTS: In 23 out of 39 patients, plasma-induced in vitro inhibition of r-αGAL A activity was observed (inhibition-positive). The inhibition titer was strongly negatively correlated to the decrease in lysoGb3: agalsidase-alfa (FElog10(inhibition) = -10.3, P ≤.001), agalsidase-beta (FElog10(inhibition) = -4.7, P ≤.001). Inhibition-positive patients had an accelerated decline in renal function (FE = 1.21, p = .042). During treatment IgG1 anti-r-αGAL A levels increased only in inhibition-positive patients (p = .0045). IgG4 anti-r-αGAL A antibodies developed in 7 out of 9 inhibition-positive patients. Other antibody subclasses were either not present or too low to quantify. CONCLUSION: Development of inhibiting antibodies against r-αGAL A negatively affects the biochemical response to ERT and resulted in an accelerated decline in renal function. The presence of IgG1 and IgG4 anti-r-αGAL A antibodies is associated with in vitro αGAL A activity inhibition.


Assuntos
Anticorpos/classificação , Doença de Fabry/tratamento farmacológico , Isoenzimas/imunologia , Proteínas Recombinantes/imunologia , alfa-Galactosidase/imunologia , Adolescente , Adulto , Anticorpos/imunologia , Seguimentos , Humanos , Imunoglobulina G/imunologia , Isoenzimas/uso terapêutico , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/uso terapêutico , Estudos Retrospectivos , Resultado do Tratamento , Adulto Jovem , alfa-Galactosidase/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA