Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Aging (Albany NY) ; 16(7): 6290-6313, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38575204

RESUMO

BACKGROUND: Immunogenic cell death (ICD) is a regulated form of cell death that triggers an adaptive immune response. The objective of this study was to investigate the correlation between ICD-related genes (ICDGs) and the prognosis and the immune microenvironment of patients with lung adenocarcinoma (LUAD). METHODS: ICD-associated molecular subtypes were identified through consensus clustering. Subsequently, a prognostic risk model comprising 5 ICDGs was constructed using Lasso-Cox regression in the TCGA training cohort and further tested in the GEO cohort. Enriched pathways among the subtypes were analyzed using GO, KEGG, and GSVA. Furthermore, the immune microenvironment was assessed using ESTIMATE, CIBERSORT, and ssGSEA analyses. RESULTS: Consensus clustering divided LUAD patients into three ICDG subtypes with significant differences in prognosis and the immune microenvironment. A prognostic risk model was constructed based on 5 ICDGs and it was used to classify the patients into two risk groups; the high-risk group had poorer prognosis and an immunosuppressive microenvironment characterized by low immune score, low immune status, high abundance of immunosuppressive cells, and high expression of tumor purity. Cox regression, ROC curve analysis, and a nomogram indicated that the risk model was an independent prognostic factor. The five hub genes were verified by TCGA database, cell sublocalization immunofluorescence analysis, IHC images and qRT-PCR, which were consistent with bioinformatics analysis. CONCLUSIONS: The molecular subtypes and a risk model based on ICDGs proposed in our study are both promising prognostic classifications in LUAD, which may provide novel insights for developing accurate targeted cancer therapies.


Assuntos
Adenocarcinoma de Pulmão , Morte Celular Imunogênica , Imunoterapia , Neoplasias Pulmonares , Microambiente Tumoral , Humanos , Microambiente Tumoral/imunologia , Microambiente Tumoral/genética , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/imunologia , Adenocarcinoma de Pulmão/patologia , Adenocarcinoma de Pulmão/terapia , Adenocarcinoma de Pulmão/mortalidade , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Neoplasias Pulmonares/mortalidade , Prognóstico , Morte Celular Imunogênica/genética , Regulação Neoplásica da Expressão Gênica , Biomarcadores Tumorais/genética , Perfilação da Expressão Gênica , Masculino , Transcriptoma , Feminino
2.
Cell Rep Med ; 4(10): 101206, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37769655

RESUMO

Immunophenotyping of the tumor microenvironment (TME) is essential for enhancing immunotherapy efficacy. However, strategies for characterizing the TME exhibit significant heterogeneity. Here, we show that endoplasmic reticular oxidoreductase-1α (ERO1A) mediates an immune-suppressive TME and attenuates the response to PD-1 blockade. Ablation of ERO1A in tumor cells substantially incites anti-tumor T cell immunity and promotes the efficacy of aPD-1 in therapeutic models. Single-cell RNA-sequencing analyses confirm that ERO1A correlates with immunosuppression and dysfunction of CD8+ T cells along anti-PD-1 treatment. In human lung cancer, high ERO1A expression is associated with a higher risk of recurrence following neoadjuvant immunotherapy. Mechanistically, ERO1A ablation impairs the balance between IRE1α and PERK signaling activities and induces lethal unfolded protein responses in tumor cells undergoing endoplasmic reticulum stress, thereby enhancing anti-tumor immunity via immunogenic cell death. These findings reveal how tumor ERO1A induces immunosuppression, highlighting its potential as a therapeutic target for cancer immunotherapy.


Assuntos
Estresse do Retículo Endoplasmático , Morte Celular Imunogênica , Neoplasias Pulmonares , Glicoproteínas de Membrana , Oxirredutases , Proteínas Serina-Treonina Quinases , Humanos , Linfócitos T CD8-Positivos , Estresse do Retículo Endoplasmático/genética , Endorribonucleases/genética , Endorribonucleases/metabolismo , Morte Celular Imunogênica/genética , Oxirredutases/genética , Microambiente Tumoral , Glicoproteínas de Membrana/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/terapia , Imunoterapia
3.
Front Immunol ; 12: 781466, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34868055

RESUMO

Immunogenic cell death (ICD) has been classified as a form of regulated cell death (RCD) that is sufficient to activate an adaptive immune response. Accumulating evidence has demonstrated the ability of ICD to reshape the tumor immune microenvironment through the emission of danger signals or DAMPs, which may contribute to the immunotherapy. Currently, identification of ICD-associated biomarkers that stratify patients according to their benefit from ICD immunotherapy would be of great advantage. Here, we identified two ICD-associated subtypes by consensus clustering. ICD-high subtype was associated with the favorable clinical outcomes, abundant immune cell infiltration, and high activity of immune response signaling. Besides, we established and validated an ICD-related prognostic model that predicted the survival of HNSCC and was associated with tumor immune microenvironment. In conclusion, we established a new classification system of HNSCC based on ICD signatures. This stratification had significant clinical outcomes for estimating prognosis, as well as the immunotherapy of HNSCC patients.


Assuntos
Biomarcadores Tumorais , Morte Celular Imunogênica , Carcinoma de Células Escamosas de Cabeça e Pescoço/etiologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/mortalidade , Biologia Computacional/métodos , Análise Mutacional de DNA , Bases de Dados Genéticas , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Ontologia Genética , Humanos , Morte Celular Imunogênica/genética , Imuno-Histoquímica , Mutação , Prognóstico , Carcinoma de Células Escamosas de Cabeça e Pescoço/diagnóstico , Carcinoma de Células Escamosas de Cabeça e Pescoço/terapia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
4.
Sci Rep ; 11(1): 12264, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34112882

RESUMO

Surgery alone or combined with chemo- and/or radiation therapy remains the primary treatment for gastric cancer (GC) to date and immunotherapeutic tools such as monoclonal antibodies are only slowly being implemented. This is partly due to the fact that the immune microenvironment in GC during chemoradiation and other treatment modalities is still poorly understood. 7 gastric cancer (GC) cell lines were tested for their response to chemoradiation using 5-FU in combination with X-ray irradiation. We conducted flow cytometric analysis to determine the cells' ability to undergo immunogenic cell death (ICD) and their expression of the two immunosuppressive proteins programmed death-ligand 1 (PD-L1) and galectin-9 (Gal-9). We evaluated the overall immunogenicity of two cell lines (MKN7, MKN74) in co-culture experiments with human monocyte-derived dendritic cells (Mo-DCs). Chemoradiation induces distinct responses in different GC cell lines. We observe ICD in vitro in all tested GC cell lines in the form of calreticulin (CRT) translocation to the plasma membrane. As a resistance mechanism, these cells also upregulated Gal-9 and PD-L1. Mo-DC maturation experiments showed that GCs provoked the maturation of Mo-DCs after chemoradiation in vitro. The addition of α-PD-L1 blocking antibody further enhanced the immunogenicity of these cells while improving DC viability. Blocking Tim-3, as the main receptor for Gal-9, had no such effect. Our findings suggest that the benefits of chemoradiation can substantially depend on tumor subtype and these benefits can be offset by induced immune evasion in GC. Combination treatment using checkpoint inhibitors could potentially lead to enhanced immune responses and yield better patient outcomes.


Assuntos
Antígeno B7-H1/genética , Galectinas/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Gástricas/genética , Apoptose/genética , Antígeno B7-H1/metabolismo , Linhagem Celular Tumoral , Quimiorradioterapia , Estresse do Retículo Endoplasmático , Galectinas/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Morte Celular Imunogênica/genética , Transporte Proteico , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
5.
Cell Death Dis ; 12(4): 314, 2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33762577

RESUMO

Chemotherapeutic agents have been linked to immunogenic cell death (ICD) induction that is capable of augmenting anti-tumor immune surveillance. The cardiac glycoside oleandrin, which inhibits Na+/K+-ATPase pump (NKP), has been shown to suppress breast cancer growth via inducing apoptosis. In the present study, we showed that oleandrin treatment triggered breast cancer cell ICD by inducing calreticulin (CRT) exposure on cell surface and the release of high-mobility group protein B1 (HMGB1), heat shock protein 70/90 (HSP70/90), and adenosine triphosphate (ATP). The maturation and activation of dendritic cells (DCs) were increased by co-culturing with the oleandrin-treated cancer cells, which subsequently enhanced CD8+ T cell cytotoxicity. Murine breast cancer cell line EMT6 was engrafted into BALB/c mice, and tumor-bearing mice were administered with oleandrin intraperitoneally every day. Oleandrin inhibited tumor growth and increased tumor infiltrating lymphocytes including DCs and T cells. Furthermore, the differential mRNA expression incurred by oleandrin was investigated by mRNA sequencing and subsequently confirmed by quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting. Mechanistically, oleandrin induced endoplasmic reticulum (ER) stress-associated, caspase-independent ICD mainly through PERK/elF2α/ATF4/CHOP pathway. Pharmacological and genetic inhibition of protein kinase R-like ER kinase (PERK) suppressed oleandrin-triggered ICD. Taken together, our findings showed that oleandrin triggered ER stress and induced ICD-mediated immune destruction of breast cancer cells. Oleandrin combined with immune checkpoint inhibitors might improve the efficacy of immunotherapy.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Neoplasias da Mama/tratamento farmacológico , Cardenolídeos/uso terapêutico , Glicosídeos Cardíacos/uso terapêutico , Morte Celular Imunogênica/genética , Animais , Neoplasias da Mama/patologia , Cardenolídeos/farmacologia , Glicosídeos Cardíacos/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Camundongos
6.
Pharmacol Res ; 163: 105265, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33129983

RESUMO

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related morbidity and mortality; it has been reported that immune cell infiltration is a prognosis factor. Here we identified genes that associated with tumor immune cell infiltrate; the underlying mechanism was verified by in vivo and in vitro experiment. In this study, Weighted correlation network analysis (WGCNA) and CIBERSORT tool were used to identify MTIF2 as the hub tumor immune infiltrating gene in HCC. To investigate the underlying role played by MTIF2, MTIF2 was knocked down by transfection of shRNA targeting MTIF2, CCK8, and EdU incorporation assay was used to evaluate the effect of MTIF2 on proliferation, wound heal assay and transwell assay was used to confirm its effect on cell migration. Ecto-calreticulin on the cell surface was evaluated by flow cytometry, ATP, and HMGB1 secretion were tested to the investigated effect of MTIF2 on the immunogenic cell death (ICD) process. We found that down-regulation of MTIF2 impaired proliferation and migration capacity of HCC cells, chemoresistance to 5-Fluorouracil (5-FU) weakened after MTIF2 was knocked down. Reduced release of damage-associated molecular patterns (DAMP) was observed after MTIF2 was overexpressed, which subsequently impaired dendritic cell (DC) maturation and proliferation of CD8 + T cells. Mechanically, the co-IP experiment confirmed that MTIF2 could interact with AIFM1, prevents AIFM1 induced transcription of caspase3, and finally suppress apoptosis. In vivo experiment also used to confirm our previously conclusion, our result indicated that MTIF2 overexpression suppresses tumor apoptosis and immune cell activity in the 5-FU therapy in vivo model, by suppression maturation of tumor-infiltrated DC. Collectively, our study confirmed that MTIF2 impair drug-induced immunogenic cell death in hepatocellular carcinoma cells.


Assuntos
Carcinoma Hepatocelular/genética , Fatores de Iniciação em Eucariotos/genética , Morte Celular Imunogênica/genética , Neoplasias Hepáticas/genética , Proteínas Mitocondriais/genética , Idoso , Animais , Antimetabólitos Antineoplásicos , Apoptose , Fator de Indução de Apoptose/metabolismo , Carcinoma Hepatocelular/metabolismo , Caspase 3/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Citocinas/metabolismo , Regulação para Baixo , Fatores de Iniciação em Eucariotos/metabolismo , Feminino , Fluoruracila , Humanos , Imunossupressores , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Proteínas Mitocondriais/metabolismo , Prognóstico
7.
Trends Cancer ; 7(3): 226-239, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33199193

RESUMO

The tumor-suppressor protein p53 is mutated in approximately half of all cancers, whereas the p53 signaling network is perturbed in almost all cancers. In response to different stress stimuli, p53 selectively activates genes to elicit a cell survival or cell death response. How p53 makes the decision between life and death remains a fascinating question and an exciting field of research. Understanding how this decision is made has major implications for improving cancer treatments, particularly in recently evolved immune checkpoint inhibition therapy. We highlight progress and challenges in understanding the mechanisms governing the p53 life and death decision-making process, and discuss how this decision is relevant to immune system regulation. Finally, we discuss how knowledge of the p53 pro-survival and pro-death decision node can be applied to optimize immune checkpoint inhibitor therapy for cancer treatment.


Assuntos
Inibidores de Checkpoint Imunológico/farmacologia , Morte Celular Imunogênica/efeitos dos fármacos , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Proteína Supressora de Tumor p53/metabolismo , Animais , Antígenos de Neoplasias/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Morte Celular Imunogênica/genética , Camundongos , MicroRNAs/metabolismo , Mutação , Neoplasias/genética , Neoplasias/imunologia , Processamento de Proteína Pós-Traducional , Receptores de Antígenos Quiméricos/imunologia , Proteína Supressora de Tumor p53/genética
8.
J Immunother Cancer ; 8(1)2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32209603

RESUMO

Cells succumbing to stress via regulated cell death (RCD) can initiate an adaptive immune response associated with immunological memory, provided they display sufficient antigenicity and adjuvanticity. Moreover, multiple intracellular and microenvironmental features determine the propensity of RCD to drive adaptive immunity. Here, we provide an updated operational definition of immunogenic cell death (ICD), discuss the key factors that dictate the ability of dying cells to drive an adaptive immune response, summarize experimental assays that are currently available for the assessment of ICD in vitro and in vivo, and formulate guidelines for their interpretation.


Assuntos
Morte Celular Imunogênica/genética , Biologia Molecular/métodos , Consenso , Guias como Assunto , Humanos
9.
ACS Appl Mater Interfaces ; 12(14): 16018-16030, 2020 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-32192326

RESUMO

Blocking immune checkpoints with monoclonal antibody has been verified to achieve potential clinical successes for cancer immunotherapy. However, its application has been impeded by the "cold" tumor microenvironment. Here, weak acidity-responsive nanoparticles co-loaded with CRISPR/Cas9 and paclitaxel (PTX) with the ability to convert "cold" tumor into "hot" tumor are reported. The nanoparticles exhibited high cargo packaging capacity, superior transfection efficiency, well biocompatibility, and effective tumor accumulation. The CRISPR/Cas9 encapsulated in nanoparticles could specifically knock out cyclin-dependent kinase 5 gene to significantly attenuate the expression of programmed death-ligand 1 on tumor cells. More importantly, PTX co-delivered in nanoparticles could significantly induce immunogenic cell death, reduce regulatory T lymphocytes, repolarize tumor-associated macrophages, and enhance antitumor immunity. Therefore, the nanoparticles could effectively convert cold tumor into hot tumor, achieve effective tumor growth inhibition, and prolong overall survival from 16 to 36 days. This research provided a referable strategy for the development of combinatorial immunotherapy and chemotherapy.


Assuntos
Proliferação de Células/efeitos dos fármacos , Melanoma Experimental/imunologia , Nanopartículas/química , Paclitaxel/farmacologia , Ácidos/química , Animais , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/imunologia , Linhagem Celular Tumoral , Quinase 5 Dependente de Ciclina/genética , Humanos , Morte Celular Imunogênica/efeitos dos fármacos , Morte Celular Imunogênica/genética , Melanoma Experimental/genética , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Nanopartículas/uso terapêutico , Paclitaxel/química , Paclitaxel/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Transfecção , Microambiente Tumoral/efeitos dos fármacos
10.
J Cell Mol Med ; 24(7): 4286-4297, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32100392

RESUMO

Oncolytic Newcastle disease virus (NDV) induces immunogenic cell death (ICD), liberating danger-associated molecular patterns (DAMPs) that provokes defiance in neoplastic malignancy. The present study aims to investigate whether and how oncolytic NDV triggers ICD in prostate cancer cells. We show that NDV/FMW, an oncolytic NDV strain FMW, elicited the expression and release of several ICD markers, that is calreticulin (CRT), heat shock proteins (HSP70/90) and high-mobility group box 1 (HMGB1), in prostate cancer cells. Furthermore, pharmacological repression of apoptosis, necroptosis, autophagy or endoplasmic reticulum (ER) stress exerted diverse effects on the HMGB1 and HSP70/90 evacuation in NDV/FMW-infected prostate cancer cells. Moreover, ICD markers induced in prostate cancer cells upon NDV/FMW infection, were enhanced by either treatment with a STAT3 (signal transducer and activator of transcription 3) inhibitor or shRNA-mediated knockdown of STAT3. In nude mice bearing prostate cancer cell-derived tumours, the tumours injected with the supernatants of NDV/FMW-infected cells grew smaller than mock-treated tumours. These results indicate that oncolytic NDV provokes the expression of ICD makers in prostate cancer cells. Our data also suggest that a combination of inhibition of STAT3 with oncolytic NDV could boost NDV-based anti-tumour effects against prostate cancer.


Assuntos
Morte Celular Imunogênica/genética , Terapia Viral Oncolítica , Neoplasias da Próstata/genética , Fator de Transcrição STAT3/genética , Animais , Apoptose/genética , Autofagia/genética , Calreticulina/genética , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/genética , Regulação Neoplásica da Expressão Gênica/genética , Proteína HMGB1/genética , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP90/genética , Humanos , Masculino , Camundongos , Camundongos Nus , Necroptose/genética , Vírus da Doença de Newcastle/genética , Vírus Oncolíticos/genética , Neoplasias da Próstata/terapia , Neoplasias da Próstata/virologia , Fator de Transcrição STAT3/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
11.
JCI Insight ; 4(18)2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31534051

RESUMO

The roles of macrophages in orchestrating innate immunity through phagocytosis and T lymphocyte activation have been extensively investigated. Much less understood is the unexpected role of macrophages in direct tumor regression. Tumoricidal macrophages can indeed manifest cancer immunoediting activity in the absence of adaptive immunity. We investigated direct macrophage cytotoxicity in malignant pleural mesothelioma, a lethal cancer that develops from mesothelial cells of the pleural cavity after occupational asbestos exposure. In particular, we analyzed the cytotoxic activity of mouse RAW264.7 macrophages upon cell-cell contact with autologous AB1/AB12 mesothelioma cells. We show that macrophages killed mesothelioma cells by oxeiptosis via a mechanism involving enhancer of zeste homolog 2 (EZH2), a histone H3 lysine 27-specific (H3K27-specific) methyltransferase of the polycomb repressive complex 2 (PRC2). A selective inhibitor of EZH2 indeed impaired RAW264.7-directed cytotoxicity and concomitantly stimulated the PD-1 immune checkpoint. In the immunocompetent BALB/c model, RAW264.7 macrophages pretreated with the EZH2 inhibitor failed to control tumor growth of AB1 and AB12 mesothelioma cells. Blockade of PD-1 engagement restored macrophage-dependent antitumor activity. We conclude that macrophages can be directly cytotoxic for mesothelioma cells independent of phagocytosis. Inhibition of the PRC2 EZH2 methyltransferase reduces this activity because of PD-1 overexpression. Combination of PD-1 blockade and EZH2 inhibition restores macrophage cytotoxicity.


Assuntos
Comunicação Celular/imunologia , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Neoplasias Pulmonares/imunologia , Macrófagos/imunologia , Mesotelioma/imunologia , Receptor de Morte Celular Programada 1/imunologia , Animais , Antineoplásicos Imunológicos/farmacologia , Antineoplásicos Imunológicos/uso terapêutico , Técnicas de Cultura de Células , Linhagem Celular Tumoral/transplante , Técnicas de Cocultura , Modelos Animais de Doenças , 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 , Humanos , Morte Celular Imunogênica/efeitos dos fármacos , Morte Celular Imunogênica/genética , Neoplasias Pulmonares/terapia , Macrófagos/metabolismo , Macrófagos/transplante , Masculino , Mesotelioma/terapia , Mesotelioma Maligno , Camundongos , Ácido Peroxinitroso/metabolismo , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Células RAW 264.7/transplante , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo
12.
Int J Mol Sci ; 20(14)2019 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-31340541

RESUMO

Apoptotic cell death is usually a response to the cell's microenvironment. In the kidney, apoptosis contributes to parenchymal cell loss in the course of acute and chronic renal injury, but does not trigger an inflammatory response. What distinguishes necrosis from apoptosis is the rupture of the plasma membrane, so necrotic cell death is accompanied by the release of unprocessed intracellular content, including cellular organelles, which are highly immunogenic proteins. The relative contribution of apoptosis and necrosis to injury varies, depending on the severity of the insult. Regulated cell death may result from immunologically silent apoptosis or from immunogenic necrosis. Recent advances have enhanced the most revolutionary concept of regulated necrosis. Several modalities of regulated necrosis have been described, such as necroptosis, ferroptosis, pyroptosis, and mitochondrial permeability transition-dependent regulated necrosis. We review the different modalities of apoptosis, necrosis, and regulated necrosis in kidney injury, focusing particularly on evidence implicating cell death in ectopic renal calcification. We also review the evidence for the role of cell death in kidney injury, which may pave the way for new therapeutic opportunities.


Assuntos
Injúria Renal Aguda/metabolismo , Proteínas Reguladoras de Apoptose/genética , Calcinose/metabolismo , Células Epiteliais/metabolismo , Rim/metabolismo , Necrose/metabolismo , Traumatismo por Reperfusão/metabolismo , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/genética , Injúria Renal Aguda/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteínas Reguladoras de Apoptose/classificação , Proteínas Reguladoras de Apoptose/metabolismo , Calcinose/genética , Calcinose/patologia , Calcinose/prevenção & controle , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Ferroptose/efeitos dos fármacos , Ferroptose/genética , Regulação da Expressão Gênica , Humanos , Morte Celular Imunogênica/efeitos dos fármacos , Morte Celular Imunogênica/genética , Rim/efeitos dos fármacos , Rim/patologia , Necrose Dirigida por Permeabilidade Transmembrânica da Mitocôndria/efeitos dos fármacos , Necrose Dirigida por Permeabilidade Transmembrânica da Mitocôndria/genética , Necroptose/efeitos dos fármacos , Necroptose/genética , Necrose/genética , Necrose/patologia , Necrose/prevenção & controle , Substâncias Protetoras/farmacologia , Piroptose/efeitos dos fármacos , Piroptose/genética , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia
13.
Mol Carcinog ; 58(10): 1754-1769, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31215708

RESUMO

We have previously shown that nearly half of mesothelioma patients have tumors with low autophagy and that these patients have a significantly worse outcome than those with high autophagy. We hypothesized that autophagy may be beneficial by facilitating immunogenic cell death (ICD) of tumor cells following chemotherapy. An important hallmark of ICD is that death of tumor cells is preceded or accompanied by the release of damage-associated molecular pattern molecules (DAMPs), which then can stimulate an antitumor immune response. Therefore, we measured how autophagy affected the release of three major DAMPs: high mobility group box 1 (HMGB1), ATP, and calreticulin following chemotherapy. We found that autophagy in three-dimensional (3D) models with low autophagy at baseline could be upregulated with the cell-permeant Tat-BECN1 peptide and confirmed that autophagy in 3D models with high autophagy at baseline could be inhibited with MRT 68921 or ATG7 RNAi, as we have previously shown. In in vitro 3D spheroids, we found that, when autophagy was high or upregulated, DAMPs were released following chemotherapy; however, when autophagy was low or inhibited, DAMPs release was significantly impaired. Similarly, in ex vivo tumors, when autophagy was high or upregulated, HMGB1 was released following chemotherapy but, when autophagy was low, HMGB1 release was not seen. We conclude that autophagy can be upregulated in at least some tumors with low autophagy and that upregulation of autophagy can restore the release of DAMPs following chemotherapy. Autophagy may be necessary for ICD in this tumor.


Assuntos
Autofagia/genética , Proteína HMGB1/genética , Morte Celular Imunogênica/genética , Mesotelioma/tratamento farmacológico , Trifosfato de Adenosina/genética , Alarminas/genética , Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Proteína 7 Relacionada à Autofagia/antagonistas & inibidores , Proteínas Relacionadas à Autofagia/genética , Proteína Beclina-1/genética , Calreticulina/genética , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imunidade Celular/genética , Mesotelioma/genética , Mesotelioma/patologia , Interferência de RNA , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/patologia
14.
J Cell Mol Med ; 23(8): 4854-4865, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31210425

RESUMO

In the tumour microenvironment (TME), immunogenic cell death (ICD) plays a major role in stimulating the dysfunctional antitumour immune system. Chronic exposure of damage-associated molecular patterns (DAMPs) attracts receptors and ligands on dendritic cells (DCs) and activates immature DCs to transition to a mature phenotype, which promotes the processing of phagocytic cargo in DCs and accelerates the engulfment of antigenic components by DCs. Consequently, via antigen presentation, DCs stimulate specific T cell responses that kill more cancer cells. The induction of ICD eventually results in long-lasting protective antitumour immunity. Through the exploration of ICD inducers, recent studies have shown that there are many novel modalities with the ability to induce immunogenic cancer cell death. In this review, we mainly discussed and summarized the emerging methods for inducing immunogenic cancer cell death. Concepts and molecular mechanisms relevant to antitumour effects of ICD are also briefly discussed.


Assuntos
Antineoplásicos/uso terapêutico , Células Dendríticas/imunologia , Morte Celular Imunogênica/genética , Neoplasias/imunologia , Neoplasias/terapia , Linfócitos T/imunologia , Microambiente Tumoral/imunologia , Animais , Antineoplásicos/farmacologia , Calreticulina/genética , Calreticulina/metabolismo , Terapia Combinada , Estresse do Retículo Endoplasmático/imunologia , Humanos , Imunoterapia , Membranas Mitocondriais/imunologia , Membranas Mitocondriais/metabolismo , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Fototerapia , Microambiente Tumoral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA