Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
J Cell Biol ; 223(3)2024 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-38329462

RESUMO

TNF signaling does not result in cell death unless multiple inhibitory signals are overcome, which can be accomplished by simultaneous signaling through IFNγ. In this issue, Deng and colleagues (http://doi.org/10.1083/jcb.202305026) dissect the mechanisms by which IFNγ signaling combines with TNF to mediate cell death through caspase-8, discussed by James E. Vince.


Assuntos
Morte Celular , Interferon gama , Transdução de Sinais , Interferon gama/fisiologia , Caspase 8/fisiologia , Fatores de Necrose Tumoral/fisiologia
2.
PLoS Biol ; 19(8): e3001304, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34437534

RESUMO

Tumor necrosis factor receptor-1 (TNFR1) signaling, apart from its pleiotropic functions in inflammation, plays a role in embryogenesis as deficiency of varieties of its downstream molecules leads to embryonic lethality in mice. Caspase-8 noncleavable receptor interacting serine/threonine kinase 1 (RIPK1) mutations occur naturally in humans, and the corresponding D325A mutation in murine RIPK1 leads to death at early midgestation. It is known that both the demise of Ripk1D325A/D325A embryos and the death of Casp8-/- mice are initiated by TNFR1, but they are mediated by apoptosis and necroptosis, respectively. Here, we show that the defects in Ripk1D325A/D325A embryos occur at embryonic day 10.5 (E10.5), earlier than that caused by Casp8 knockout. By analyzing a series of genetically mutated mice, we elucidated a mechanism that leads to the lethality of Ripk1D325A/D325A embryos and compared it with that underlies Casp8 deletion-mediated lethality. We revealed that the apoptosis in Ripk1D325A/D325A embryos requires a scaffold function of RIPK3 and enzymatically active caspase-8. Unexpectedly, caspase-1 and caspase-11 are downstream of activated caspase-8, and concurrent depletion of Casp1 and Casp11 postpones the E10.5 lethality to embryonic day 13.5 (E13.5). Moreover, caspase-3 is an executioner of apoptosis at E10.5 in Ripk1D325A/D325A mice as its deletion extends life of Ripk1D325A/D325A mice to embryonic day 11.5 (E11.5). Hence, an unexpected death pathway of TNFR1 controls RIPK1 D325A mutation-induced lethality at E10.5.


Assuntos
Caspase 8/fisiologia , Desenvolvimento Embrionário , Proteína Serina-Treonina Quinases de Interação com Receptores/fisiologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Animais , Caspases/metabolismo , Morte Celular , Camundongos , Cultura Primária de Células , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo
3.
Cancer Sci ; 112(7): 2664-2678, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33934451

RESUMO

Immunotherapy targeting the PD-L1/PD-1 pathway is a novel type of clinical cancer treatment, but only small subsets of patients can benefit from it because of multiple factors. PD-L1/PD-1 expression is a biomarker for predicting the efficacy of anti-PD-L1/PD-1 therapy, which highlights the importance of understanding the regulatory mechanisms of PD-L1 expression in cancer cells. Casp8 is an apical caspase protease involved in mediating cell apoptosis, but it also has multiple nonapoptotic functions. Casp8 mutations are associated with increased risks of cancer, and low expression of Casp8 is closely connected with poor prognosis in patients with cancer. In addition, mutations of Casp8 in lymphocytes also lead to human immunodeficiency, thereby causing dysfunction of the innate immune system, but the roles of Casp8 in antitumor immunity remain unclear. Here, we found that knocking down Casp8 in mouse melanoma cells promoted tumor progression in an immune system-dependent manner. Mechanistically, Casp8 induced PD-L1 degradation by upregulating TNFAIP3 (A20) expression, a ubiquitin-editing enzyme that results in PD-L1 ubiquitination. In addition, compared with Casp8fl/fl mice, mice with conditional deletion of Casp8 in natural killer (NK) cells (Ncr1iCre/+ Casp8fl/fl mice) showed a decreased frequency of IFN-γ+ and CD107a+ NK cells but an increased frequency of PD-1+ and CTLA-4+ NK cells. Melanoma cells with Casp8 knocked down exhibited sensitivity to anti-PD-1 or anti-CTLA-4 antibody treatments, particularly in Ncr1iCre/+Casp8fl/fl mice. Together, the results indicate that Casp8 induces PD-L1 degradation by upregulating A20 expression and that decreased Casp8 expression is a potential biomarker for predicting the sensitivity to anti-PD-L1/PD-1 immunotherapy.


Assuntos
Antígeno B7-H1/metabolismo , Caspase 8/fisiologia , Imunoterapia Adotiva/métodos , Melanoma/terapia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , Animais , Antígeno B7-H1/genética , Antígeno CTLA-4/metabolismo , Caspase 8/genética , Linhagem Celular Tumoral , Progressão da Doença , Regulação para Baixo , Proteínas Ativadoras de GTPase/metabolismo , Interferon gama/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Proteínas de Membrana Lisossomal/metabolismo , Melanoma/imunologia , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , NF-kappa B/metabolismo , Ubiquitinação , Regulação para Cima
4.
Cancer Metastasis Rev ; 40(1): 303-318, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33026575

RESUMO

Caspase-8 is an aspartate-specific cysteine protease, which is best known for its apoptotic functions. Caspase-8 is placed at central nodes of multiple signal pathways, regulating not only the cell cycle but also the invasive and metastatic cell behavior, the immune cell homeostasis and cytokine production, which are the two major components of the tumor microenvironment (TME). Ovarian cancer often has dysregulated caspase-8 expression, leading to imbalance between its apoptotic and non-apoptotic functions within the tumor and the surrounding milieu. The downregulation of caspase-8 in ovarian cancer seems to be linked to high aggressiveness with chronic inflammation, immunoediting, and immune resistance. Caspase-8 plays therefore an essential role not only in the primary tumor cells but also in the TME by regulating the immune response, B and T lymphocyte activation, and macrophage differentiation and polarization. The switch between M1 and M2 macrophages is possibly associated with changes in the caspase-8 expression. In this review, we are discussing the non-apoptotic functions of caspase-8, highlighting this protein as a modulator of the immune response and the cytokine composition in the TME. Considering the low survival rate among ovarian cancer patients, it is urgently necessary to develop new therapeutic strategies to optimize the response to the standard treatment. The TME is highly heterogenous and provides a variety of opportunities for new drug targets. Given the variety of roles of caspase-8 in the TME, we should focus on this protein in the development of new therapeutic strategies against the TME of ovarian cancer.


Assuntos
Caspase 8/fisiologia , Neoplasias Ovarianas , Microambiente Tumoral , Feminino , Humanos , Macrófagos , Transdução de Sinais
5.
JCI Insight ; 5(23)2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33108350

RESUMO

Caspase-8 (CASP8) is one of the most frequently mutated genes in head and neck squamous carcinomas (HNSCCs), and CASP8 mutations are associated with poor survival. The distribution of these mutations in HNSCCs suggests that they are likely to be inactivating. Inhibition of CASP8 has been reported to sensitize cancer cells to necroptosis, a regulated cell death mechanism. Here, we show that knockdown of CASP8 renders HNSCCs susceptible to necroptosis by a second mitochondria-derived activator of caspase (SMAC) mimetic, birinapant, in combination with pan-caspase inhibitors Z-VAD-FMK or emricasan and radiation. In a syngeneic mouse model of oral cancer, birinapant, particularly when combined with radiation, delayed tumor growth and enhanced survival under CASP8 loss. Exploration of molecular underpinnings of necroptosis sensitivity confirmed that the level of functional receptor-interacting serine/threonine protein kinase 3 (RIP3) determines susceptibility to this mode of death. Although an in vitro screen revealed that low RIP3 levels rendered many HNSCC cell lines resistant to necroptosis, patient tumors maintained RIP3 expression and should therefore remain sensitive. Collectively, these results suggest that targeting the necroptosis pathway with SMAC mimetics, especially in combination with radiation, may be relevant therapeutically in HNSCC with compromised CASP8 status, provided that RIP3 function is maintained.


Assuntos
Caspase 8/metabolismo , Necroptose/fisiologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Biomimética , Caspase 8/genética , Caspase 8/fisiologia , Inibidores de Caspase/metabolismo , Inibidores de Caspase/farmacologia , Caspases/metabolismo , Linhagem Celular Tumoral , Bases de Dados Genéticas , Dipeptídeos/metabolismo , Dipeptídeos/farmacologia , Humanos , Indóis/metabolismo , Indóis/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Necroptose/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética
6.
Cell Rep ; 32(4): 107959, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32726624

RESUMO

Pyroptosis has emerged as a key mechanism by which inflammasomes promote host defense against microbial pathogens and sterile inflammation. Gasdermin D (GSDMD)-mediated cell lysis is a hallmark of pyroptosis, but our understanding of cell death signaling during pyroptosis is fragmented. Here, we show that independently of GSDMD-mediated plasma membrane permeabilization, inflammasome receptors engage caspase-1 and caspase-8, both of which redundantly promote activation of apoptotic executioner caspase-3 and caspase-7 in pyroptotic macrophages. Impaired GSDMD pore formation downstream of caspase-1 and caspase-8 activation suffices to unmask the apoptotic phenotype of pyroptotic macrophages. Combined inactivation of initiator caspase-1 and caspase-8, or executioner caspase-3 and caspase-7, is required to abolish inflammasome-induced DEVDase activity during pyroptosis and in apoptotic Gsdmd-/- cells. Collectively, these results unveil a robust apoptotic caspase network that is activated in parallel to GSDMD-mediated plasma membrane permeabilization and safeguards cell death induction in pyroptotic macrophages.


Assuntos
Caspases/metabolismo , Macrófagos/metabolismo , Piroptose/fisiologia , Animais , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/metabolismo , Caspase 1/metabolismo , Caspase 1/fisiologia , Caspase 3/metabolismo , Caspase 7/metabolismo , Caspase 8/metabolismo , Caspase 8/fisiologia , Morte Celular , Membrana Celular/metabolismo , Feminino , Inflamassomos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Ligação a Fosfato/metabolismo
7.
Am J Physiol Gastrointest Liver Physiol ; 317(4): G493-G507, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31411503

RESUMO

Genetically modified mice have been successfully used as models for inflammatory bowel diseases; however, dietary effects were poorly examined. Here, we studied the impact of particular nutrients and supplements on gut functions related to the knockout of the epithelial caspase-8 gene. Caspase-8 knockout (Casp8∆IEC) and control (Casp8fl) mice were fed for 4 wk a control diet (CD) enriched with 10% inulin (CD-Inu) or 5% sodium butyrate (CD-But) while having free access to plain water or water supplemented with 30% fructose (+F). Body weight changes, intestinal inflammation, and selected markers for barrier function and of liver steatosis were assessed. Casp8∆IEC mice developed ileocolitis accompanied by changes in intestinal barrier morphology and reduced expression of barrier-related genes such as mucin-2 (Muc2) and defensins in the ileum and Muc2 in the colon. Casp8∆IEC mice fed a CD also showed impaired body weight gain compared with Casp8fl mice, which was even more pronounced in mice receiving water supplemented with fructose. Furthermore, we observed a marked liver steatosis and inflammation in some but not all Casp8∆IEC mice under a CD, which was on average similar to that observed in control mice under a fructose-rich diet. Hepatic lipid accumulation, as well as markers of ileal barrier function, but not intestinal pathohistology or body weight loss, were attenuated by diets enriched with inulin or butyrate, especially in the absence of fructose supplementation. Our data show that ileocolitis, barrier dysfunction, and malassimilation in Caspase-8 knockout mice can be partially attenuated by oral inulin or butyrate supplementation.NEW & NOTEWORTHY Genetic mouse models for ileocolitis are important to understand inflammatory bowel disease in humans. We examined dietetic factors that might aggravate or attenuate ileocolitis and related pathologies in such a model. Deletion of the caspase-8 gene results not only in ileocolitis but also in gut barrier dysfunction, liver steatosis, and malassimilation, which can be partially attenuated by oral inulin or sodium butyrate. Our data indicate that diet modifications can contribute to disease variability and therapy.


Assuntos
Ácido Butírico/farmacologia , Caspase 8/genética , Caspase 8/fisiologia , Doença de Crohn/genética , Doença de Crohn/patologia , Mucosa Intestinal/patologia , Inulina/farmacologia , Animais , Peso Corporal/genética , Doença de Crohn/tratamento farmacológico , Dieta Ocidental , Suplementos Nutricionais , Feminino , Microbioma Gastrointestinal , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucina-2/genética , Mucina-2/metabolismo , Nutrientes
8.
Mol Nutr Food Res ; 63(12): e1801206, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30900802

RESUMO

SCOPE: Pyrrolizidine alkaloids (PAs) are common phytotoxins. Intoxication can lead to liver damage. Previous studies showed PA-induced apoptosis in liver cells. However, the exact role of the extrinsic apoptotic pathway has not been investigated yet. This study aims to analyze whether the PA representative lasiocarpine sensitizes human liver cells toward extrinsic Fas-mediated apoptosis. METHODS AND RESULTS: HepG2 cells with limited xenobiotic metabolic activity are used to analyze metabolism-dependent effects. External in vitro metabolism is simulated using rat or human liver enzymes. Additionally, metabolically competent HepaRG cells are used to confirm the observed effects in a human liver cell system with internal xenobiotic metabolism. Metabolized lasiocarpine decreases cell viability and induces Fas receptor gene expression in both cell lines. Increased Fas receptor protein expression on the cell surface is demonstrated by flow cytometry. The addition of a Fas ligand-simulating antibody induces apoptosis. Induction of extrinsic Fas-mediated apoptosis is verified by Western blotting for cleaved caspase 8, the initiator caspase of extrinsic apoptosis. All effects are dependent on lasiocarpine metabolism. CONCLUSION: The results demonstrate that metabolically metabolized lasiocarpine sensitizes human liver cells toward Fas-mediated apoptosis. They broaden our knowledge on the hepatotoxic molecular mechanisms of PA as widely distributed food contaminants.


Assuntos
Apoptose/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Alcaloides de Pirrolizidina/farmacologia , Receptor fas/fisiologia , Ativação Metabólica , Animais , Caspase 8/fisiologia , Proteína Ligante Fas/farmacologia , Células Hep G2 , Hepatócitos/fisiologia , Humanos , Masculino , Alcaloides de Pirrolizidina/farmacocinética , Ratos , Ratos Wistar
9.
BMC Vet Res ; 15(1): 20, 2019 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-30621683

RESUMO

BACKGROUND: Caspases are a family of conserved intracellular cysteine-dependent aspartate-specific cysteine proteases that play important roles in regulating cell death and inflammation. Our previous study revealed the importance of the inflammatory caspase 1 gene in extracellular ATP-mediated immune signaling in Japanese flounder, Paralichthys olivaceus. To explore the potential roles of other caspases in P. olivaceus innate immunity, we extended our study by characterizing of the responses of four additional P. olivaceus caspase genes, termed JfCaspase 2, 3, 6 and 8, to inflammatory challenge and extracellular ATP stimulation. RESULTS: Sequence analysis revealed that the domain structures of all the Japanese flounder caspase proteins are evolutionarily conserved. Quantitative real-time PCR analysis showed that the JfCaspase 2, 3, 6 and 8 genes were expressed ubiquitously but at unequal levels in all examined Japanese flounder normal tissues. In addition, the basal gene expression levels of JfCaspase 2, 3, 6 and 8 were higher than those of JfCaspase 1 in both Japanese flounder head kidney macrophages (HKMs) and peripheral blood leukocytes (PBLs). Furthermore, immune challenge experiments showed that the inflammatory stimuli LPS and poly(I:C) significantly modulated the expression of the JfCaspase 2, 3, 6 and 8 genes in Japanese flounder immune cells. Finally, DNA fragmentation, associated with increased extracellular ATP-induced JfCaspase 2, 3, 6 and 8 gene expression and enzymatic activity, was inhibited by the caspase inhibitor Z-VAD-FMK in the HKMs. CONCLUSION: Our findings demonstrate broad participation of multiple caspase genes in response to inflammatory stimulation in Japanese flounder immune cells and provide new evidence for the involvement of caspase(s) in extracellular ATP-induced apoptosis in fish.


Assuntos
Trifosfato de Adenosina/farmacologia , Caspase 2/genética , Caspase 3/genética , Caspase 6/genética , Caspase 8/genética , Proteínas de Peixes/genética , Linguado/imunologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Caspase 2/fisiologia , Caspase 3/fisiologia , Caspase 6/fisiologia , Caspase 8/fisiologia , Proteínas de Peixes/fisiologia , Linguado/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Genes/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Lipopolissacarídeos/farmacologia , Filogenia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Alinhamento de Sequência/veterinária , Análise de Sequência de DNA/veterinária
10.
Med Sci Monit ; 24: 5247-5257, 2018 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-30057402

RESUMO

BACKGROUND Periodontal ligament stem cells (PDLSCs) possess characteristics of multi-potential differentiation and immuno-modulation, and PDLSCs-mediated periodontal tissue regeneration is regarded as a hopeful method for periodontitis treatment. Recent studies demonstrated that RIP3 and caspase8 regulate bacteria-induced innate immune response and programmed necrosis, which is also called necroptosis. This study aimed to determine the role of the RIP3/Caspase8 signal pathway on necroptosis of PDLSCs under the inflammatory microenvironment, both [i]in vitro[/i] and [i]in vivo[/i]. MATERIAL AND METHODS PDLSCs were cultured, and transmission electron microscopy and flow cytometry were used to detect necroptosis. PCR, ALP, and Alizarin Red S staining were used to assess the effect of necroptosis on osteogenesis differentiation of PDLSCs [i]in vitro[/i], while HE and Masson staining were taken after the nude mouse subcutaneous transplant experiment. RESULTS Our research indicates that RIP3/caspase8 can regulate the immune response of PDLSCs, and blockade of RIP3/caspase8 can protect the biological characteristics of the PDLSCs, effectively promoting periodontal tissue regeneration in the inflammatory microenvironment. CONCLUSIONS Inhibiting RIP3/caspase8 can effectively promote periodontal tissue regeneration in the inflammatory microenvironment.


Assuntos
Caspase 8/fisiologia , Periodontite/terapia , Proteína Serina-Treonina Quinases de Interação com Receptores/fisiologia , Animais , Caspase 8/metabolismo , Diferenciação Celular/fisiologia , China , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Necrose/fisiopatologia , Osteogênese/efeitos dos fármacos , Ligamento Periodontal/citologia , Ligamento Periodontal/fisiologia , Periodontite/fisiopatologia , Cultura Primária de Células , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Regeneração/fisiologia , Transdução de Sinais/fisiologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/citologia , Células-Tronco/fisiologia
11.
Sci Rep ; 8(1): 3788, 2018 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-29491424

RESUMO

The NLRC4 inflammasome recognizes bacterial flagellin and components of the type III secretion apparatus. NLRC4 stimulation leads to caspase-1 activation followed by a rapid lytic cell death known as pyroptosis. NLRC4 is linked to pathogen-free auto-inflammatory diseases, suggesting a role for NLRC4 in sterile inflammation. Here, we show that NLRC4 activates an alternative cell death program morphologically similar to apoptosis in caspase-1-deficient BMDMs. By performing an unbiased genome-wide CRISPR/Cas9 screen with subsequent validation studies in gene-targeted mice, we highlight a critical role for caspase-8 and ASC adaptor in an alternative apoptotic pathway downstream of NLRC4. Furthermore, caspase-1 catalytically dead knock-in (Casp1 C284A KI) BMDMs genetically segregate pyroptosis and apoptosis, and confirm that caspase-1 does not functionally compete with ASC for NLRC4 interactions. We show that NLRC4/caspase-8-mediated apoptotic cells eventually undergo plasma cell membrane damage in vitro, suggesting that this pathway can lead to secondary necrosis. Unexpectedly, we found that DFNA5/GSDME, a member of the pore-forming gasdermin family, is dispensable for the secondary necrosis that follows NLRC4-mediated apoptosis in macrophages. Together, our data confirm the existence of an alternative caspase-8 activation pathway diverging from the NLRC4 inflammasome in primary macrophages.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Proteínas Adaptadoras de Sinalização CARD/fisiologia , Proteínas de Ligação ao Cálcio/metabolismo , Caspase 1/fisiologia , Caspase 8/fisiologia , Inflamassomos/metabolismo , Macrófagos/patologia , Animais , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/genética , Sistemas CRISPR-Cas , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/genética , Genoma , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
Biochim Biophys Acta Gen Subj ; 1862(3): 557-566, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29203282

RESUMO

BACKGROUND: The development of approaches that increase therapeutic effects of anti-cancer drugs is one of the most important tasks of oncology. Caloric restriction in vivo or serum deprivation (SD) in vitro has been shown to be an effective tool for sensitizing cancer cells to chemotherapeutic drugs. However, the detailed mechanisms underlying the enhancement of apoptosis in cancer cells by SD remain to be elucidated. METHODS: Flow cytometry, caspase activity assay and western blotting were used for cell death rate evaluation. Western blotting, gel-filtration, siRNA approach and qRT-PCR were used to elucidate the mechanism underlying cell death potentiation upon SD. RESULTS: We demonstrated that SD sensitizes cancer cells to treatment with chemotherapeutic agent cisplatin. This effect is independent on activation of caspases-2 and -8, apical caspases triggering apoptosis in response to genotoxic stress. SD potentiates cell death via downregulation of the anti-apoptotic protein Mcl-1. In fact, SD reduces the Mcl-1 mRNA level, which consequently decreases the Mcl-1 protein level and renders cells more susceptible to apoptosis induction via the formation of apoptosome. CONCLUSIONS: Mcl-1 protein is an important regulator of sensitivity of cancer cells to apoptotic stimuli upon SD. GENERAL SIGNIFICANCE: This study identifies Mcl-1 as a new target for the sensitization of human cancer cells to cell death by SD, which is of great significance for the development of efficient anti-cancer therapies.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Apoptose/efeitos dos fármacos , Cisplatino/farmacologia , Meios de Cultura Livres de Soro/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteína de Sequência 1 de Leucemia de Células Mieloides/biossíntese , Proteínas de Neoplasias/biossíntese , Apoptose/fisiologia , Apoptossomas/fisiologia , Caspase 2/fisiologia , Caspase 8/fisiologia , Linhagem Celular Tumoral , Cisteína Endopeptidases/fisiologia , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Células HeLa , Humanos , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides/fisiologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Interferência de RNA , RNA Interferente Pequeno/genética
13.
Oncogene ; 36(31): 4434-4444, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28368418

RESUMO

In the process of cancer spreading, different modes of invasion exist. One is expansive invasion, in which a group of cancer cells gradually expands along with cancer cell proliferation. Invasion of cancer cells is also modified by their interaction with stromal cells including cancer-associated fibroblasts (CAFs). Cancer cells co-invade with CAFs, and invasion by CAFs frequently precede invasion by cancer cells, which indicates CAF-led cancer cell invasion. Here, we show that CAFs induce apoptosis in gastric cancer cells, which prevented expansive invasion by cancer cells and instead facilitated CAF-led invasion. Death receptor 4 and activation of caspase-8 in cancer cells mediated cancer cell apoptosis induced by CAFs, which was dependent on contact between cancer cells and CAFs. Apoptotic cancer cells in turn released apoptotic vesicles and stimulated invasion of CAFs. Accordingly, cancer cells followed the migrating CAFs. Treatment with a caspase inhibitor, ZVAD, or forced expression of a death domain fragment in cancer cells prevented cancer cell apoptosis induced by CAFs and increased expansive invasion by cancer cells in extracellular gel invasion assays, while the rate of cancer cell invasion led by CAFs was decreased. Death domain-fragment expression also prevented intramural invasion by gastric cancer cells in the stomach. Because CAF-led invasion is characterized by the movement of individual cancer cells away from the tumour, adequate cancer cell apoptosis may promote cancer dissemination.


Assuntos
Apoptose , Fibroblastos Associados a Câncer/fisiologia , Invasividade Neoplásica , Neoplasias Gástricas/patologia , Animais , Caspase 8/fisiologia , Comunicação Celular , Linhagem Celular Tumoral , Vesículas Extracelulares/fisiologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/fisiologia
14.
Sci Rep ; 6: 29041, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27383564

RESUMO

Glomerular mesangial cell (GMC) proliferation and death are involved in the pathogenesis of glomerular disorders. The mechanisms that control GMC survival are poorly understood, but may include signal transduction pathways that are modulated by changes in intracellular Ca(2+) ([Ca(2+)]i) concentration. In this study, we investigated whether activation of the canonical transient receptor potential (TRPC) 6 channels and successive [Ca(2+)]i elevation alter neonatal GMC survival. Hyperforin (HF)-induced TRPC6 channel activation increased [Ca(2+)]i concentration, inhibited proliferation, and triggered apoptotic cell death in primary neonatal pig GMCs. HF-induced neonatal GMC apoptosis was not associated with oxidative stress. However, HF-induced TRPC6 channel activation stimulated nuclear translocation of the nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1). HF also increased cell death surface receptor Fas ligand (FasL) level and caspase-8 activity in the cells; effects mitigated by [Ca(2+)]i chelator BAPTA, calcineurin/NFAT inhibitor VIVIT, and TRPC6 channel knockdown. Accordingly, HF-induced neonatal GMC apoptosis was attenuated by BAPTA, VIVIT, Fas blocking antibody, and a caspase-3/7 inhibitor. These findings suggest that TRPC6 channel-dependent [Ca(2+)]i elevation and the ensuing induction of the calcineurin/NFAT, FasL/Fas, and caspase signaling cascades promote neonatal pig GMC apoptosis.


Assuntos
Apoptose/fisiologia , Calcineurina/fisiologia , Sinalização do Cálcio/fisiologia , Proteína Ligante Fas/fisiologia , Transporte de Íons/fisiologia , Células Mesangiais/metabolismo , Fatores de Transcrição NFATC/fisiologia , Transdução de Sinais/fisiologia , Canal de Cátion TRPC6/fisiologia , Receptor fas/fisiologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Cálcio/farmacologia , Sinalização do Cálcio/genética , Caspase 8/fisiologia , Inibidores de Caspase/farmacologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Transporte de Íons/efeitos dos fármacos , Células Mesangiais/efeitos dos fármacos , Oligopeptídeos/farmacologia , Estresse Oxidativo , Floroglucinol/análogos & derivados , Floroglucinol/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Sus scrofa , Canal de Cátion TRPC6/antagonistas & inibidores , Canal de Cátion TRPC6/genética , Terpenos/farmacologia
15.
FEBS J ; 283(14): 2616-25, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26395133

RESUMO

Necroptosis is a form of programmed cell death that is both mechanistically and morphologically distinct from apoptosis, the canonical mechanism of cell suicide. Although early descriptions of necroptosis date back decades, the last 5 years have seen a proliferation of studies of this process. This surge in interest has included the recent publication of several excellent, in-depth reviews of the literature [Chan FK-M et al. (2014) Annu Rev Immunol 33, 141210135520002; Weinlich R & Green DR (2014) Mol Cell 56, 469-480; Silke J et al. (2015) Nat Immunol 16, 689-697; Linkermann A & Green DR (2014) N Engl J Med 370, 455-465]. Rather than contribute another summary to this well-summarized field, in this Minireview I will briefly discuss key recent findings, then touch on some of the major outstanding questions - the known unknowns - that remain.


Assuntos
Morte Celular/fisiologia , Alarminas/fisiologia , Animais , Apoptose/imunologia , Apoptose/fisiologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/fisiologia , Caspase 8/fisiologia , Morte Celular/imunologia , Humanos , Proteínas Inibidoras de Apoptose/fisiologia , Modelos Biológicos , Necrose , Neoplasias/patologia , Neoplasias/fisiopatologia , Moléculas com Motivos Associados a Patógenos/metabolismo , Proteínas Quinases/fisiologia , Proteína Serina-Treonina Quinases de Interação com Receptores/fisiologia
16.
J Appl Toxicol ; 35(6): 603-13, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25639670

RESUMO

The purpose of this study was to determine the intracellular signaling transduction pathways involved in oxidative stress induced by nanoparticles in cancer cells. Activation of reactive oxygen species (ROS) has some therapeutic benefits in arresting the growth of cancer cells. Cobalt oxide nanoparticles (CoO NPs) are an interesting compound for oxidative cancer therapy. Our results showed that CoO NPs elicited a significant (P <0.05) amount of ROS in cancer cells. Co-treatment with N-aceyltine cystine (an inhibitor of ROS) had a protective role in cancer cell death induced by CoO NPs. In cultured cells, the elevated level of tumor necrosis factor-alpha (TNF-α) was noted after CoO NPs treatment. This TNF-α persuaded activation of caspase-8 followed by phosphorylation of p38 mitogen-activated protein kinase and induced cell death. This study showed that CoO NPs induced oxidative stress and activated the signaling pathway of TNF-α-Caspase-8-p38-Caspase-3 to cancer cells.


Assuntos
Caspase 8/fisiologia , Cobalto/efeitos adversos , Leucemia Experimental/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Nanopartículas Metálicas/efeitos adversos , Estresse Oxidativo/efeitos dos fármacos , Óxidos/efeitos adversos , Fator de Necrose Tumoral alfa/fisiologia , Acetilcisteína/farmacologia , Animais , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Microscopia Eletrônica de Transmissão , Transplante de Neoplasias , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier
17.
Cancer Res ; 75(6): 986-95, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25600646

RESUMO

Immune mechanisms underlying hepatocellular carcinoma (HCC) are not well understood. Here, we show that the Toll-like receptor TLR2 inhibits production of the proinflammatory cytokine IL18 and protects mice from DEN-induced liver carcinogenesis. On this protocol, Tlr2(-/-) mice exhibited more aggressive HCC development associated with impaired CD8(+) T-cell function. Furthermore, Ly6C(high)IL18Rα(+) myeloid-derived suppressor cells (MDSC) were increased in number in the livers of Tlr2(-/-) mice before tumor onset. MDSC in this setting exhibited higher iNOS levels that could inhibit IFNγ production and CD8(+) T-cell proliferation in vitro. Notably, Tlr2(-/-) hepatocytes produced more mature IL18 after DEN treatment that was sufficient to drive MDSC accumulation there. IL18 administration was sufficient to induce accumulation of MDSC, whereas hepatocyte-specific silencing of IL18 in Tlr2(-/-) mice decreased the proportion of MDSC, increased the proportion of functional CD8(+) T cells, and alleviated HCC progression. IL18 production was mediated by caspase-8 insofar as the decrease in its silencing was sufficient to attenuate levels of mature IL18 in Tlr2(-/-) mice. Furthermore, the TLR2 agonist Pam3CSK4 inhibited both caspase-8 and IL18 expression, decreasing MDSC, increasing CD8(+) T-cell function, and promoting HCC regression. Overall, our findings show how TLR2 deficiency accelerates IL18-mediated immunosuppression during liver carcinogenesis, providing new insights into immune control that may assist the design of effective immunotherapies to treat HCC.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Tolerância Imunológica , Interleucina-18/fisiologia , Neoplasias Hepáticas Experimentais/etiologia , Receptor 2 Toll-Like/fisiologia , Animais , Caspase 8/fisiologia , Neoplasias Hepáticas Experimentais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/fisiologia
18.
Cell Cycle ; 14(7): 1070-81, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25602147

RESUMO

Stathmin/Oncoprotein 18, a microtubule destabilizing protein, is required for survival of p53-deficient cells. Stathmin-depleted cells are slower to enter mitosis, but whether delayed mitotic entry triggers cell death or whether stathmin has a separate pro-survival function was unknown. To test these possibilities, we abrogated the cell cycle delay by inhibiting Wee1 in synchronized, stathmin-depleted cells and found that apoptosis was reduced to control levels. Synchronized cells treated with a 4 hour pulse of inhibitors to CDK1 or both Aurora A and PLK1 delayed mitotic entry and apoptosis was triggered only in p53-deficient cells. We did not detect mitotic defects downstream of the delayed mitotic entry, indicating that cell death is activated by a mechanism distinct from those activated by prolonged mitotic arrest. Cell death is triggered by initiator caspase 8, based on its cleavage to the active form and by rescue of viability after caspase 8 depletion or treatment with a caspase 8 inhibitor. In contrast, initiator caspase 9, activated by prolonged mitotic arrest, is not activated and is not required for apoptosis under our experimental conditions. P53 upregulates expression of cFLIPL, a protein that blocks caspase 8 activation. cFLIPL levels are lower in cells lacking p53 and these levels are reduced to a greater extent after stathmin depletion. Expression of FLAG-tagged cFLIPL in p53-deficient cells rescues them from apoptosis triggered by stathmin depletion or CDK1 inhibition during G2. These data indicate that a cell cycle delay in G2 activates caspase 8 to initiate apoptosis specifically in p53-deficient cells.


Assuntos
Caspase 8/fisiologia , Mitose , Proteína Supressora de Tumor p53/metabolismo , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Caspase 9/metabolismo , Pontos de Checagem da Fase G2 do Ciclo Celular , Células HCT116 , Células HeLa , Humanos , Estatmina/metabolismo
19.
Gut ; 64(4): 601-10, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25379949

RESUMO

OBJECTIVES: Intestinal epithelial cells (IEC) express toll-like receptors (TLR) that facilitate microbial recognition. Stimulation of TLR ligands induces a transient increase in epithelial cell shedding, a mechanism that serves the antibacterial and antiviral host defence of the epithelium and promotes elimination of intracellular pathogens. Although activation of the extrinsic apoptosis pathway has been described during inflammatory shedding, its functional involvement is currently unclear. DESIGN: We investigated the functional involvement of caspase-8 signalling in microbial-induced intestinal cell shedding by injecting Lipopolysaccharide (LPS) to mimic bacterial pathogens and poly(I:C) as a probe for RNA viruses in vivo. RESULTS: TLR stimulation of IEC was associated with a rapid activation of caspase-8 and increased epithelial cell shedding. In mice with an epithelial cell-specific deletion of caspase-8 TLR stimulation caused Rip3-dependent epithelial necroptosis instead of apoptosis. Mortality and tissue damage were more severe in mice in which IECs died by necroptosis than apoptosis. Inhibition of receptor-interacting protein (Rip) kinases rescued the epithelium from TLR-induced gut damage. TLR3-induced necroptosis was directly mediated via TRIF-dependent pathways, independent of Tnf-α and type III interferons, whereas TLR4-induced tissue damage was critically dependent on Tnf-α. CONCLUSIONS: Together, our data demonstrate an essential role for caspase-8 in maintaining the gut barrier in response to mucosal pathogens by permitting inflammatory shedding and preventing necroptosis of infected cells. These data suggest that therapeutic strategies targeting the cell death machinery represent a promising new option for the treatment of inflammatory and infective enteropathies.


Assuntos
Caspase 8/fisiologia , Células Epiteliais/fisiologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Camundongos , Transdução de Sinais
20.
Circulation ; 130(24): 2162-72, 2014 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-25278099

RESUMO

BACKGROUND: Programmed necrosis (necroptosis) plays an important role in development, tissue homeostasis, and disease pathogenesis. The molecular mechanisms that regulate necroptosis in the heart and its physiological relevance in myocardial remodeling and heart failure remain largely unknown. METHODS AND RESULTS: Here, we identified an obligate function for TAK1 (transforming growth factor ß-activated kinase 1, gene name Map3k7) in regulating necroptotic myocyte death, myocardial remodeling, and heart failure propensity. Cardiac-specific ablation of Map3k7 in mice induced spontaneous apoptosis and necroptosis that led to adverse remodeling and heart failure, and these effects were abolished by ablation of tumor necrosis factor receptor-1. Mechanistically, TAK1 functions as a molecular switch in tumor necrosis factor receptor-1 signaling by regulating the formation of 2 cell death complexes, RIP 1 (receptor-interacting protein 1)-FADD (Fas-associated protein with death domain)-caspase 8 and RIP1-RIP3, a process that is dependent on FADD and caspase 8 as scaffolding molecules. Importantly, inhibition of RIP1 or RIP3 largely blocked necroptotic cell death, adverse remodeling, and heart failure in TAK1-deficient mice. CONCLUSIONS: These results indicate that TAK1 functions as a key survival factor in the heart by directly antagonizing necroptosis, which is critical for the maintenance of myocardial homeostasis and the prevention of adverse myocardial remodeling.


Assuntos
Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , MAP Quinase Quinase Quinases/fisiologia , Transdução de Sinais/fisiologia , Remodelação Ventricular/fisiologia , Animais , Apoptose/fisiologia , Caspase 8/fisiologia , Linhagem Celular , Modelos Animais de Doenças , Proteína de Domínio de Morte Associada a Fas/fisiologia , Insuficiência Cardíaca/mortalidade , Homeostase/fisiologia , MAP Quinase Quinase Quinases/deficiência , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Necrose/fisiopatologia , Proteína Serina-Treonina Quinases de Interação com Receptores/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA