Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 176(6): 1447-1460.e14, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30799039

RESUMEN

The presence of DNA in the cytoplasm is normally a sign of microbial infections and is quickly detected by cyclic GMP-AMP synthase (cGAS) to elicit anti-infection immune responses. However, chronic activation of cGAS by self-DNA leads to severe autoimmune diseases for which no effective treatment is available yet. Here we report that acetylation inhibits cGAS activation and that the enforced acetylation of cGAS by aspirin robustly suppresses self-DNA-induced autoimmunity. We find that cGAS acetylation on either Lys384, Lys394, or Lys414 contributes to keeping cGAS inactive. cGAS is deacetylated in response to DNA challenges. Importantly, we show that aspirin can directly acetylate cGAS and efficiently inhibit cGAS-mediated immune responses. Finally, we demonstrate that aspirin can effectively suppress self-DNA-induced autoimmunity in Aicardi-Goutières syndrome (AGS) patient cells and in an AGS mouse model. Thus, our study reveals that acetylation contributes to cGAS activity regulation and provides a potential therapy for treating DNA-mediated autoimmune diseases.


Asunto(s)
ADN/inmunología , Nucleotidiltransferasas/metabolismo , Autotolerancia/inmunología , Acetilación , Secuencia de Aminoácidos , Animales , Aspirina/farmacología , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Autoinmunidad , Línea Celular , ADN/genética , ADN/metabolismo , Modelos Animales de Enfermedad , Exodesoxirribonucleasas/metabolismo , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Mutación , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/inmunología , Malformaciones del Sistema Nervioso/metabolismo , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/química , Nucleotidiltransferasas/genética , Células THP-1
2.
Nat Immunol ; 20(1): 18-28, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30510222

RESUMEN

Cyclic GMP-AMP synthase (cGAS) is a key sensor responsible for cytosolic DNA detection. Here we report that GTPase-activating protein SH3 domain-binding protein 1 (G3BP1) is critical for DNA sensing and efficient activation of cGAS. G3BP1 enhanced DNA binding of cGAS by promoting the formation of large cGAS complexes. G3BP1 deficiency led to inefficient DNA binding by cGAS and inhibited cGAS-dependent interferon (IFN) production. The G3BP1 inhibitor epigallocatechin gallate (EGCG) disrupted existing G3BP1-cGAS complexes and inhibited DNA-triggered cGAS activation, thereby blocking DNA-induced IFN production both in vivo and in vitro. EGCG administration blunted self DNA-induced autoinflammatory responses in an Aicardi-Goutières syndrome (AGS) mouse model and reduced IFN-stimulated gene expression in cells from a patient with AGS. Thus, our study reveals that G3BP1 physically interacts with and primes cGAS for efficient activation. Furthermore, EGCG-mediated inhibition of G3BP1 provides a potential treatment for cGAS-related autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , ADN Helicasas/metabolismo , Complejos Multiproteicos/metabolismo , Malformaciones del Sistema Nervioso/metabolismo , Nucleotidiltransferasas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Animales , Autoantígenos/inmunología , Autoantígenos/metabolismo , Enfermedades Autoinmunes del Sistema Nervioso/tratamiento farmacológico , Enfermedades Autoinmunes del Sistema Nervioso/genética , Catequina/análogos & derivados , Catequina/uso terapéutico , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Citosol/inmunología , Citosol/metabolismo , ADN/inmunología , ADN/metabolismo , ADN Helicasas/antagonistas & inhibidores , ADN Helicasas/genética , Modelos Animales de Enfermedad , Exodesoxirribonucleasas/genética , Células HEK293 , Células HeLa , Humanos , Interferones/metabolismo , Ratones , Ratones Noqueados , Malformaciones del Sistema Nervioso/tratamiento farmacológico , Malformaciones del Sistema Nervioso/genética , Fármacos Neuroprotectores/uso terapéutico , Fosfoproteínas/genética , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores , Proteínas de Unión a Poli-ADP-Ribosa/genética , Unión Proteica , ARN Helicasas/antagonistas & inhibidores , ARN Helicasas/genética , Proteínas con Motivos de Reconocimiento de ARN/antagonistas & inhibidores , Proteínas con Motivos de Reconocimiento de ARN/genética
3.
Breast Cancer Res ; 25(1): 10, 2023 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-36703228

RESUMEN

Necroptosis is a form of regulated necrosis and is executed by MLKL when MLKL is engaged in triggering the rupture of cell plasma membrane. MLKL activation also leads to the protease, ADAMs-mediated ectodomain shedding of cell surface proteins of necroptotic cells. Tumor necroptosis often happens in advanced solid tumors, and blocking necroptosis by MLKL deletion in breast cancer dramatically reduces tumor metastasis. It has been suggested that tumor necroptosis affects tumor progression through modulating the tumor microenvironment. However, the exact mechanism by which tumor necroptosis promotes tumor metastasis remains elusive. Here, we report that the ectodomain shedding of cell surface proteins of necroptotic cells is critical for the promoting effect of tumor necroptosis in tumor metastasis through inhibiting the anti-tumor activity of T cells. We found that blocking tumor necroptosis by MLKL deletion led to the dramatic reduction of tumor metastasis and significantly elevated anti-tumor activity of tumor-infiltrating and peripheral blood T cells. Importantly, the increased anti-tumor activity of T cells is a key cause for the reduced metastasis as the depletion of CD8+ T cells completely restored the level of metastasis in the Mlkl KO mice. Interestingly, the levels of some soluble cell surface proteins including sE-cadherin that are known to promote metastasis are also dramatically reduced in MLKL null tumors/mice. Administration of ADAMs pan inhibitor reduces the levels of soluble cell surface proteins in WT tumors/mice and leads to the dramatic decrease in metastasis. Finally, we showed the sE-cadherin/KLRG1 inhibitory receptor is the major pathway for necroptosis-mediated suppression of the anti-tumor activity of T cells and the promotion of metastasis. Hence, our study reveals a novel mechanism of tumor necroptosis-mediated promotion of metastasis and suggests that tumor necroptosis and necroptosis-activated ADAMs are potential targets for controlling metastasis.


Asunto(s)
Neoplasias de la Mama , Proteínas de la Membrana , Necroptosis , Metástasis de la Neoplasia , Animales , Ratones , Cadherinas , Proteínas de la Membrana/metabolismo , Ratones Noqueados , Proteínas Quinasas , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/farmacología , Microambiente Tumoral , Neoplasias de la Mama/patología
4.
Nat Immunol ; 11(9): 799-805, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20711193

RESUMEN

MicroRNAs are key regulators of many biological processes, including cell differentiation. Here we show that during human monocyte-macrophage differentiation, expression of the microRNAs miR-223, miR-15a and miR-16 decreased considerably, which led to higher expression of the serine-threonine kinase IKKalpha in macrophages. In macrophages, higher IKKalpha expression in conjunction with stabilization of the kinase NIK induced larger amounts of p52. Because of low expression of the transcription factor RelB in untreated macrophages, high p52 expression repressed basal transcription of both canonical and noncanonical NF-kappaB target genes. However, proinflammatory stimuli in macrophages resulted in greater induction of noncanonical NF-kappaB target genes. Thus, a decrease in certain microRNAs probably prevents macrophage hyperactivation yet primes the macrophage for certain responses to proinflammatory stimuli.


Asunto(s)
Diferenciación Celular/inmunología , Regulación del Desarrollo de la Expresión Génica , Quinasa I-kappa B/inmunología , Quinasa I-kappa B/metabolismo , Macrófagos/inmunología , MicroARNs/inmunología , FN-kappa B/inmunología , Células Cultivadas , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Macrófagos/citología , FN-kappa B/genética , Transducción de Señal , Células U937 , Regulación hacia Arriba
5.
Mol Cell ; 42(5): 597-609, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21658601

RESUMEN

The regulation of apoptosis is critical for controlling tissue homeostasis and preventing tumor formation and growth. Reactive oxygen species (ROS) generation plays a key role in such regulation. Here, we describe a HIF-1 target, Vasn/ATIA (anti-TNFα-induced apoptosis), which protects cells against TNFα- and hypoxia-induced apoptosis. Through the generation of ATIA knockout mice, we show that ATIA protects cells from apoptosis through regulating the function of the mitochondrial antioxidant, thioredoxin-2, and ROS generation. ATIA is highly expressed in human glioblastoma, and ATIA knockdown in glioblastoma cells renders them sensitive to hypoxia-induced apoptosis. Therefore, ATIA is not only a HIF-1 target that regulates mitochondrial redox pathways but also a potentially diagnostic marker and therapeutic target in human glioblastoma.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Apoptosis , Proteínas Portadoras/fisiología , Factor 1 Inducible por Hipoxia/metabolismo , Proteínas de la Membrana/fisiología , Tiorredoxinas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/análisis , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Portadoras/química , Proteínas Portadoras/genética , Hipoxia de la Célula , Línea Celular Tumoral , Membrana Celular/metabolismo , Glioblastoma/metabolismo , Humanos , Proteínas de la Membrana/análisis , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Mitocondrias/metabolismo , Oxidación-Reducción , Tiorredoxinas/genética
6.
J Biol Chem ; 291(38): 20030-41, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27489105

RESUMEN

NADPH oxidases (NOXs) are involved in inflammation, angiogenesis, tumor growth, and osteoclast differentiation. However, the role of NOX1 and NOX2 in macrophage differentiation and tumor progression is still elusive. Here we report that NOX1 and NOX2 are critical for the differentiation of monocytes to macrophages, the polarization of M2-type but not M1-type macrophages, and the occurrence of tumor-associated macrophages (TAMs). We found that deletion of both NOX1 and NOX2 led to a dramatic decrease in ROS production in macrophages and resulted in impaired efficiency in monocyte-to-macrophage differentiation and M2-type macrophage polarization. We further showed that NOX1 and NOX2 were critical for the activation of the MAPKs JNK and ERK during macrophage differentiation and that the deficiency of JNK and ERK activation was responsible for the failure of monocyte-to-macrophage differentiation, in turn affecting M2 macrophage polarization. Furthermore, we demonstrated that the decrease in M2 macrophages and TAMs, concomitant with the reduction of cytokine and chemokine secretion, contributed to the delay in wound healing and the inhibition of tumor growth and metastasis in NOX1/2 double knockout mice compared with WT mice. Collectively, these data provide direct evidence that NOX1 and NOX2 deficiency impairs macrophage differentiation and the occurrence of M2-type TAMs during tumor development.


Asunto(s)
Diferenciación Celular/inmunología , Macrófagos/inmunología , Glicoproteínas de Membrana/inmunología , Monocitos/inmunología , NADH NADPH Oxidorreductasas/inmunología , NADPH Oxidasas/inmunología , Especies Reactivas de Oxígeno/inmunología , Animales , Diferenciación Celular/genética , Quimiocinas/genética , Quimiocinas/inmunología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/inmunología , Macrófagos/enzimología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Monocitos/enzimología , NADH NADPH Oxidorreductasas/genética , NADH NADPH Oxidorreductasas/metabolismo , NADPH Oxidasa 1 , NADPH Oxidasa 2 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
7.
Proc Natl Acad Sci U S A ; 109(14): 5322-7, 2012 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-22421439

RESUMEN

Tumor necrosis factor (TNF) is an important inflammatory cytokine and induces many cellular responses, including inflammation, cell proliferation, apoptosis, and necrosis. It is known that receptor interacting protein (RIP) kinases, RIP1 and RIP3, are key effectors of TNF-induced necrosis, but little is known about how these two RIP kinases mediate this process, although reactive oxygen species (ROS) generation and JNK activation have been suggested to be two downstream events of RIP kinases. Here we report the identification of mixed lineage kinase domain-like, MLKL, as a key RIP3 downstream component of TNF-induced necrosis. Through screening a kinase/phosphatase shRNA library in human colon adenocarcinoma HT-29 cells, we found that knockdown of MLKL blocked TNF-induced necrosis. Our data suggest that MLKL functions downstream of RIP1 and RIP3 and is recruited to the necrosome through its interaction with RIP3. Finally, we found that MLKL is required for the generation of ROS and the late-phase activation of JNK during TNF-induced necrosis. However, because these two events are not involved in TNF-induced necrosis in HT-29 cells, the target of MLKL during TNF-induced necrosis remains elusive. Taken together, our study suggests that MLKL is a key RIP3 downstream component of TNF-induced necrotic cell death.


Asunto(s)
Necrosis , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Secuencia de Bases , Línea Celular , Cartilla de ADN , Activación Enzimática , Humanos , MAP Quinasa Quinasa 4/metabolismo , Fosforilación , Reacción en Cadena de la Polimerasa , Especies Reactivas de Oxígeno/metabolismo
8.
Blood ; 119(12): 2895-905, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22223827

RESUMEN

Monocytes are programmed to undergo apoptosis in the absence of stimulation. Stimuli that promote monocyte-macrophage differentiation not only cause cellular changes, but also prevent the default apoptosis of monocytes. In the present study, we demonstrate that autophagy is induced when monocytes are triggered to differentiate and that the induction of autophagy is pivotal for the survival and differentiation of monocytes. We also show that inhibition of autophagy results in apoptosis of cells that are engaged in differentiation. We found that the differentiation signal releases Beclin1 from Bcl-2 by activating JNK and blocks Atg5 cleavage, both of which are critical for the induction of autophagy. Preventing autophagy induction hampers differentiation and cytokine production; therefore, autophagy is an important transition from monocyte apoptosis to differentiation.


Asunto(s)
Autofagia/fisiología , Diferenciación Celular/fisiología , Macrófagos/citología , Monocitos/citología , Animales , Western Blotting , Células Cultivadas , Citometría de Flujo , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Electrónica de Transmisión , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología
9.
J Cell Sci ; 124(Pt 4): 647-56, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21266470

RESUMEN

The current paradigm of noncanonical NF-κB signaling suggests that the loss of TRAF2, TRAF3 or cIAP1 and cIAP2 leads to stabilization of NF-κB-inducing kinase (NIK) to activate the noncanonical pathway. Although a crucial role of RIP1 in the TNFα-induced canonical NF-κB pathway has been well established, its involvement in noncanonical activation of NF-κB through the TNFR1 receptor, is unknown. Here we show that TNFα is capable of activating the noncanonical NF-κB pathway, but that activation of this pathway is negatively regulated by RIP1. In the absence of RIP1, TNFR1 stimulation leads to activation of the noncanonical NF-κB pathway through TRAF2 degradation, leading to NIK stabilization, IKKα phosphorylation and the processing of p100 to generate p52. Thus although RIP1(-/-) mouse embryonic fibroblasts are sensitive at early time points to cell death induced by TNFα, probably as a result of lack of canonical NF-κB activation, the late activation of the noncanonical NF-κB pathway protects the remaining cells from further cell death. The TNFR1-dependent noncanonical NF-κB activation in RIP1(-/-) cells suggests that there is functional interplay between the two NF-κB pathways during TNFR1 signaling, which might regulate the number and kinds of NF-κB transcription factors and thus finely control NF-κB-dependent gene transcription.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , FN-kappa B/metabolismo , Factor 2 Asociado a Receptor de TNF/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular , Proteínas Activadoras de GTPasa/genética , Humanos , Ratones , Ratones Noqueados , FN-kappa B/genética , Unión Proteica , Estabilidad Proteica , Transducción de Señal , Factor 2 Asociado a Receptor de TNF/química , Factor 2 Asociado a Receptor de TNF/genética , Factor de Necrosis Tumoral alfa/genética
10.
J Immunol ; 186(9): 5212-6, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21421854

RESUMEN

TNFR-associated death domain protein (TRADD) is a key effector protein of TNFR1 signaling. However, the role of TRADD in other death receptor (DR) signaling pathways, including DR3, has not been completely characterized. Previous studies using overexpression systems suggested that TRADD is recruited to the DR3 complex in response to the DR3 ligand, TNF-like ligand 1A (TL1A), indicating a possible role in DR3 signaling. Using T cells from TRADD knockout mice, we demonstrate in this study that the response of both CD4(+) and CD8(+) T cells to TL1A is dependent upon the presence of TRADD. TRADD knockout T cells therefore lack the appropriate proliferative response to TL1A. Moreover, in the absence of TRADD, both the stimulation of MAPK signaling and activation of NF-κB in response to TL1A are dramatically reduced. Unsurprisingly, TRADD is required for recruitment of receptor interacting protein 1 and TNFR-associated factor 2 to the DR3 signaling complex and for the ubiquitination of receptor interacting protein 1. Thus, our findings definitively establish an essential role of TRADD in DR3 signaling.


Asunto(s)
Activación de Linfocitos/inmunología , Miembro 25 de Receptores de Factores de Necrosis Tumoral/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Proteína de Dominio de Muerte Asociada a Receptor de TNF/inmunología , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Animales , Western Blotting , Separación Celular , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Inmunoprecipitación , Ratones , Ratones Noqueados , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Linfocitos T/metabolismo , Proteína de Dominio de Muerte Asociada a Receptor de TNF/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo
11.
FASEB J ; 25(4): 1353-8, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21187341

RESUMEN

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily. TRAIL is promising for anticancer therapy because it induces apoptosis in cancer cells with little or no toxicity to normal cells; hence, TRAIL-receptor agonists are currently undergoing clinical trials for cancer treatment. However, many molecular signaling mechanisms in TRAIL signaling are not completely characterized. The functions of adaptor proteins, including TNF-receptor-associated death domain protein (TRADD) and receptor-interacting protein-1 (RIP1) in TRAIL signaling have been controversial. We demonstrate that while wild-type mouse embryonic fibroblasts (MEFs) are completely resistant to TRAIL-induced apoptosis, MEFs derived from Tradd(-/-) mice are hypersensitive to TRAIL (IC(50)~0.5 nM rmTRAIL, 24 h), an effect also seen in primary keratinocytes treated with TRAIL/CHX. Restoration of TRADD in Tradd(-/-) MEFs restores TRAIL resistance, indicating that TRADD plays a survival role in TRAIL signaling. We show that TRADD is recruited to the TRAIL-receptor complex, and RIP1 recruitment is mediated by TRADD. While early activation of the MAP kinase ERK is deficient in Tradd(-/-) cells, the main mechanism for enhanced TRAIL sensitivity is likely due to increased recruitment of FADD to the receptor complex, indicating that TRADD may limit FADD binding within the receptor complex and also mediate RIP1-dependent nonapoptotic signaling events, thus reducing caspase activation and subsequent apoptosis. These novel findings have potential implications for cancer therapy using TRAIL-receptor agonists.


Asunto(s)
Transducción de Señal/fisiología , Proteína de Dominio de Muerte Asociada a Receptor de TNF/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Animales , Apoptosis/fisiología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Ratones , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/agonistas
12.
STAR Protoc ; 3(3): 101457, 2022 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-35719728

RESUMEN

Necroptosis occurs predominantly in the center of late-stage tumors and necroptotic cells are dispersed and difficult to be detected by Western blotting of key markers without enrichment by microdissection. To overcome these obstacles, this protocol provides a detailed immunohistochemistry-oriented approach including the steps of tumor isolation from mouse mammary tumor models, necrotic region identification by H&E staining, and necroptosis detection through examining mixed lineage kinase domain-like protein (MLKL) phosphorylation. This protocol could be applied to other types of solid tumors. For complete details on the use and execution of this protocol, please refer to Baik et al. (2021).


Asunto(s)
Neoplasias Mamarias Animales , Necroptosis , Animales , Ratones , Necrosis/patología , Fosforilación , Proteínas Quinasas/metabolismo , Factores de Transcripción/metabolismo
13.
Trends Cancer ; 8(1): 21-27, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34627742

RESUMEN

Necroptosis, a form of programmed necrotic cell death, is a gatekeeper of host defense against certain pathogen invasions. The deregulation of necroptosis is also a key factor of many inflammatory diseases. Recent studies have revealed an important role of necroptosis in tumorigenesis and metastasis and imply the potential of targeting necroptosis as a novel cancer therapy. While its molecular mechanism has been well studied, details of the regulation and function of necroptosis of tumor cells in tumorigenesis and metastasis only began to emerge recently, and we discuss these herein.


Asunto(s)
Necroptosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Apoptosis/genética , Humanos , Necroptosis/genética , Necrosis , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
14.
J Cell Biol ; 175(4): 607-17, 2006 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-17116751

RESUMEN

Cross talk between NF-kappaB and c-Jun N-terminal kinases (JNKs) has been implicated in the cell life and death decision under various stresses. Functional suppression of JNK activation by NF-kappaB has recently been proposed as a key cellular survival mechanism and contributes to cancer cells escaping from apoptosis. We provide a novel scenario of the proapoptotic role of IkappaB kinase beta (IKKbeta)-NF-kappaB, which can act as the activator of the JNK pathway through the induction of GADD45alpha for triggering MKK4/JNK activation, in response to the stimulation of arsenite, a cancer therapeutic reagent. This effect of IKKbeta-NF-kappaB is dependent on p50 but not the p65/relA NF-kappaB subunit, which can increase the stability of GADD45alpha protein through suppressing its ubiquitination and proteasome-dependent degradation. IKKbeta-NF-kappaB can therefore either activate or suppress the JNK cascade and consequently mediate pro- or antiapoptotic effects, depending on the manner of its induction. Furthermore, the NF-kappaB p50 subunit can exert a novel regulatory function on protein modification independent of the classical NF-kappaB transcriptional activity.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsenitos/farmacología , Proteínas de Ciclo Celular/metabolismo , Quinasa I-kappa B/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Animales , Activación Enzimática/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Quinasa I-kappa B/deficiencia , Ratones , Subunidad p50 de NF-kappa B/deficiencia , Fosforilación/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Ubiquitina/metabolismo , Regulación hacia Arriba/efectos de los fármacos
15.
J Immunol ; 183(5): 3278-84, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19641134

RESUMEN

The serine-threonine kinase RIP1 was originally identified through its ability to bind to the death domain of Fas (CD95). RIP1 has been shown to be recruited to the Fas death-inducing signaling complex (DISC) and is required for the induction of necrotic cell death. In this study, we show that in Jurkat T lymphocytes, RIP1 is also necessary for the most efficient activation of downstream caspases by Fas when treated with membrane-bound Fas ligand, but not with agonistic Abs or cross-linked soluble Fas ligand. RIP1 participates in the Fas-associated death domain protein-mediated recruitment of caspase-8 to the Fas receptor complex in a manner that promotes caspase-8 activation. Cross-linking Abs, such as CH11, bypass the requirement for RIP1 in caspase activation by initiating larger, though less efficient, DISC complexes, while membrane-bound Fas ligand initiates a smaller but more efficient DISC that functions, in part, by effectively incorporating more RIP1 into the complex. Consequently, RIP1 is likely a more integral part of physiological signaling through the Fas/CD95 receptor complex than previously recognized; at least when the signal is mediated by full-length membrane-bound FasL. Cross-linked soluble FasL, which also occurs physiologically, behaves similarly to the CH11 Ab, and may therefore be more likely to initiate nonapoptotic Fas signaling due to less RIP1 in the receptor complex. Thus, agonists that bind the same Fas receptor initiate mechanistically distinct pathways resulting in differential cytotoxicity.


Asunto(s)
Caspasa 8/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Proteína Ligando Fas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Complejo Poro Nuclear/fisiología , Proteínas de Unión al ARN/fisiología , Anticuerpos/metabolismo , Muerte Celular/inmunología , Reactivos de Enlaces Cruzados/metabolismo , Citotoxicidad Inmunológica , Activación Enzimática/inmunología , Proteína Ligando Fas/inmunología , Proteína Ligando Fas/toxicidad , Humanos , Inmunidad Innata , Inmunoglobulina M/metabolismo , Células Jurkat , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/toxicidad , Proteínas de Complejo Poro Nuclear/deficiencia , Proteínas de Complejo Poro Nuclear/metabolismo , Oligopéptidos , Péptidos/inmunología , Proteínas de Unión al ARN/metabolismo , Receptor fas/agonistas , Receptor fas/inmunología , Receptor fas/metabolismo
16.
Methods Mol Biol ; 2248: 73-80, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33185868

RESUMEN

Tumor necrosis factor (TNF) plays a key role in inflammatory responses and in various cellular events such as apoptosis and necroptosis. The interaction of TNF with its receptor, TNFR1, drives the initiation of complex molecular pathways leading to inflammation and cell death. RARγ is released from the nucleus to orchestrate the formation of the cytosolic death complexes, and it is cytosolic RARγ that plays a pivotal role in switching TNF-induced inflammatory responses to RIPK1-initiated cell death. Thus, RARγ provides a checkpoint for the transition from inflammatory signaling to death machinery of RIPK1-initiated cell death in response to TNF. Here, we use techniques to identify RARγ as a downstream mediator of TNFR1 signaling complex. We use confocal imaging to show the localization of RARγ upon activation of cell death. Immunoprecipitation of RARγ identified the interacting proteins.


Asunto(s)
Apoptosis , Inflamación/etiología , Inflamación/metabolismo , Transducción de Señal , Factores de Necrosis Tumoral/metabolismo , Animales , Apoptosis/genética , Biomarcadores , Western Blotting , Línea Celular , Susceptibilidad a Enfermedades , Humanos , Inmunoprecipitación , Inflamación/patología , Factores de Necrosis Tumoral/genética
17.
Nat Commun ; 12(1): 2666, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976222

RESUMEN

Tumor necrosis happens commonly in advanced solid tumors. We reported that necroptosis plays a major role in tumor necrosis. Although several key necroptosis regulators including receptor interacting protein kinase 1 (RIPK1) have been identified, the regulation of tumor necroptosis during tumor development remains elusive. Here, we report that Z-DNA-binding protein 1 (ZBP1), not RIPK1, mediates tumor necroptosis during tumor development in preclinical cancer models. We found that ZBP1 expression is dramatically elevated in necrotic tumors. Importantly, ZBP1, not RIPK1, deletion blocks tumor necroptosis during tumor development and inhibits metastasis. We showed that glucose deprivation triggers ZBP1-depedent necroptosis in tumor cells. Glucose deprivation causes mitochondrial DNA (mtDNA) release to the cytoplasm and the binding of mtDNA to ZBP1 to activate MLKL in a BCL-2 family protein, NOXA-dependent manner. Therefore, our study reveals ZBP1 as the key regulator of tumor necroptosis and provides a potential drug target for controlling tumor metastasis.


Asunto(s)
Neoplasias de la Mama/genética , Necroptosis/genética , Proteínas de Unión al ARN/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Células HEK293 , Humanos , Células MCF-7 , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Proteínas de Unión al ARN/metabolismo , Tratamiento con ARN de Interferencia/métodos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
18.
Mol Pharmacol ; 77(5): 784-92, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20159942

RESUMEN

Cytokine-activated inhibitor of kappaB kinase beta (IKKbeta) is a key mediator of immune and inflammatory responses, but recent studies suggest that IKKbeta is also required for tissue homeostasis in physiopathological processes. Here we report a novel role for IKKbeta in maintenance of constitutive levels of the redox scavenger GSH. Inactivation of IKKbeta by genetic or pharmacological means results in low cellular GSH content and marked reduction of redox potential. Similar to Ikkbeta(-/-) cells, Tnfr1(-/-) and p65(-/-) cells are also GSH-deficient. As a consequence, cells deficient in IKKbeta signaling are extremely susceptible to toxicity caused by environmental and pharmacological agents, including oxidants, genotoxic agents, microtubule toxins, and arsenic. GSH biosynthesis depends on the activity of the rate-limiting enzyme glutamate-cysteine ligase (GCL), consisting of a catalytic subunit (GCLC) and a modifier subunit (GCLM). We found that loss of IKKbeta signaling significantly reduces basal NF-kappaB activity and decreases binding of NF-kappaB to the promoters of Gclc and Gclm, leading to reduction of GCLC and GCLM expression. Conversely, overexpression of GCLC and GCLM in IKKbeta-null cells partially restores GSH content and prevents stress-induced cytotoxicity. We suggest that maintenance of GSH is a novel physiological role of the IKKbeta-NF-kappaB signaling cascade to prevent oxidative damage and preserve the functional integrity of the cells.


Asunto(s)
Quinasa I-kappa B/genética , FN-kappa B/genética , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Supervivencia Celular , Células Cultivadas , Cartilla de ADN , Genes Reporteros , Glutatión/deficiencia , Glutatión/genética , Glutatión/metabolismo , Homeostasis , Quinasa I-kappa B/deficiencia , Quinasa I-kappa B/metabolismo , Quinasa I-kappa B/farmacología , Luciferasas/genética , Ratones , Ratones Noqueados , Oxidación-Reducción , Plásmidos , Especies Reactivas de Oxígeno/metabolismo
19.
Cancers (Basel) ; 12(10)2020 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-33066043

RESUMEN

New approaches to target MYC include the stabilization of a guanine-rich, G-quadruplex (G4) tertiary DNA structure in the NHE III region of its promoter. Recent screening of a small molecule microarray platform identified a benzofuran, D089, that can stabilize the MYC G4 and inhibit its transcription. D089 induced both dose- and time-dependent multiple myeloma cell death mediated by endoplasmic reticulum induced stress. Unexpectedly, we uncovered two mechanisms of cell death: cellular senescence, as evidenced by increased levels of p16, p21 and γ-H2AX proteins and a caspase 3-independent mechanism consistent with pyroptosis. Cells treated with D089 exhibited high levels of the cleaved form of initiator caspase 8; but failed to show cleavage of executioner caspase 3, a classical apoptotic marker. Cotreatment with the a pan-caspase inhibitor Q-VD-OPh did not affect the cytotoxic effect of D089. In contrast, cleaved caspase 1, an inflammatory caspase downstream of caspases 8/9, was increased by D089 treatment. Cells treated with D089 in addition to either a caspase 1 inhibitor or siRNA-caspase 1 showed increased IC50 values, indicating a contribution of cleaved caspase 1 to cell death. Downstream effects of caspase 1 activation after drug treatment included increases in IL1B, gasdermin D cleavage, and HMGB1 translocation from the nucleus to the cytoplasm. Drug treated cells underwent a 'ballooning' morphology characteristic of pyroptosis, rather than 'blebbing' typically associated with apoptosis. ASC specks colocalized with NLRP3 in proximity ligation assays after drug treatment, indicating inflammasome activation and further confirming pyroptosis as a contributor to cell death. Thus, the small molecule MYC G4 stabilizer, D089, provides a new tool compound for studying pyroptosis. These studies suggest that inducing both tumor senescence and pyroptosis may have therapeutic potential for cancer treatment.

20.
Transl Oncol ; 13(1): 32-41, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31760267

RESUMEN

BACKGROUND: Escaping cell death pathways is an important event during carcinogenesis. We previously identified anti-TNFα-induced apoptosis (ATIA, also known as vasorin) as an antiapoptotic factor that suppresses reactive oxygen species (ROS) production. However, the role of vasorin in lung carcinogenesis has not been investigated. METHODS: Vasorin expression was examined in human lung cancer tissues with immunohistochemistry and database analysis. Genetic and pharmacological approaches were used to manipulate protein expression and autophagy activity in human bronchial epithelial cells (HBECs). ROS generation was measured with fluorescent indicator, apoptosis with release of lactate dehydrogenase, and cell transformation was assessed with colony formation in soft agar. RESULTS: Vasorin expression was increased in human lung cancer tissues and cell lines, which was inversely associated with lung cancer patient survival. Cigarette smoke extract (CSE) and benzo[a]pyrene diol epoxide (BPDE)-induced vasorin expression in HBECs. Vasorin knockdown in HBECs significantly suppressed CSE-induced transformation in association with enhanced ROS accumulation and autophagy. Scavenging ROS attenuated autophagy and cytotoxicity in vasorin knockdown cells, suggesting that vasorin potentiates transformation by impeding ROS-mediated CSE cytotoxicity and improving survival of the premalignant cells. Suppression of autophagy effectively inhibited CSE-induced apoptosis, suggesting that autophagy was pro-apoptotic in CSE-treated cells. Importantly, blocking autophagy strongly potentiated CSE-induced transformation. CONCLUSION: These results suggest that vasorin is a potential lung cancer-promoting factor that facilitates cigarette smoke-induced bronchial epithelial cell transformation by suppressing autophagy-mediated apoptosis, which could be exploited for lung cancer prevention.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA