Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nature ; 575(7784): 683-687, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31748744

RESUMEN

Caspase-8 is the initiator caspase of extrinsic apoptosis1,2 and inhibits necroptosis mediated by RIPK3 and MLKL. Accordingly, caspase-8 deficiency in mice causes embryonic lethality3, which can be rescued by deletion of either Ripk3 or Mlkl4-6. Here we show that the expression of enzymatically inactive CASP8(C362S) causes embryonic lethality in mice by inducing necroptosis and pyroptosis. Similar to Casp8-/- mice3,7, Casp8C362S/C362S mouse embryos died after endothelial cell necroptosis leading to cardiovascular defects. MLKL deficiency rescued the cardiovascular phenotype but unexpectedly caused perinatal lethality in Casp8C362S/C362S mice, indicating that CASP8(C362S) causes necroptosis-independent death at later stages of embryonic development. Specific loss of the catalytic activity of caspase-8 in intestinal epithelial cells induced intestinal inflammation similar to intestinal epithelial cell-specific Casp8 knockout mice8. Inhibition of necroptosis by additional deletion of Mlkl severely aggravated intestinal inflammation and caused premature lethality in Mlkl knockout mice with specific loss of caspase-8 catalytic activity in intestinal epithelial cells. Expression of CASP8(C362S) triggered the formation of ASC specks, activation of caspase-1 and secretion of IL-1ß. Both embryonic lethality and premature death were completely rescued in Casp8C362S/C362SMlkl-/-Asc-/- or Casp8C362S/C362SMlkl-/-Casp1-/- mice, indicating that the activation of the inflammasome promotes CASP8(C362S)-mediated tissue pathology when necroptosis is blocked. Therefore, caspase-8 represents the molecular switch that controls apoptosis, necroptosis and pyroptosis, and prevents tissue damage during embryonic development and adulthood.


Asunto(s)
Apoptosis/genética , Caspasa 8/genética , Caspasa 8/metabolismo , Necroptosis/genética , Piroptosis/genética , Animales , Línea Celular , Células Cultivadas , Activación Enzimática/genética , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Inflamasomas/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/enzimología , Queratinocitos/citología , Queratinocitos/patología , Ratones , Mutación , Receptor TIE-2/genética , Receptor TIE-2/metabolismo
2.
EMBO Rep ; 23(6): e53608, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35437868

RESUMEN

Elevated expression of the X-linked inhibitor of apoptosis protein (XIAP) has been frequently reported in malignant melanoma suggesting that XIAP renders apoptosis resistance and thereby supports melanoma progression. Independent of its anti-apoptotic function, XIAP mediates cellular inflammatory signalling and promotes immunity against bacterial infection. The pro-inflammatory function of XIAP has not yet been considered in cancer. By providing detailed in vitro analyses, utilising two independent mouse melanoma models and including human melanoma samples, we show here that XIAP is an important mediator of melanoma neutrophil infiltration. Neutrophils represent a major driver of melanoma progression and are increasingly considered as a valuable therapeutic target in solid cancer. Our data reveal that XIAP ubiquitylates RIPK2, involve TAB1/RIPK2 complex and induce the transcriptional up-regulation and secretion of chemokines such as IL8, that are responsible for intra-tumour neutrophil accumulation. Alteration of the XIAP-RIPK2-TAB1 inflammatory axis or the depletion of neutrophils in mice reduced melanoma growth. Our data shed new light on how XIAP contributes to tumour growth and provides important insights for novel XIAP targeting strategies in cancer.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis , Melanoma , Infiltración Neutrófila , Neoplasias Cutáneas , Proteína Inhibidora de la Apoptosis Ligada a X , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Apoptosis/genética , Apoptosis/inmunología , Modelos Animales de Enfermedad , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/inmunología , Interleucina-8/biosíntesis , Melanoma/genética , Melanoma/inmunología , Ratones , Infiltración Neutrófila/genética , Infiltración Neutrófila/inmunología , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/inmunología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Proteína Inhibidora de la Apoptosis Ligada a X/inmunología , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
3.
EMBO Rep ; 22(10): e52301, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34342114

RESUMEN

Maintaining the architecture, size and composition of an intact stem cell (SC) compartment is crucial for tissue homeostasis and regeneration throughout life. In mammalian skin, elevated expression of the anti-apoptotic Bcl-2 protein has been reported in hair follicle (HF) bulge SCs (BSCs), but its impact on SC function is unknown. Here, we show that systemic exposure of mice to the Bcl-2 antagonist ABT-199/venetoclax leads to the selective loss of suprabasal BSCs (sbBSCs), thereby disrupting cyclic HF regeneration. RNAseq analysis shows that the pro-apoptotic BH3-only proteins BIM and Bmf are upregulated in sbBSCs, explaining their addiction to Bcl-2 and the marked susceptibility to Bcl-2 antagonism. In line with these observations, conditional knockout of Bcl-2 in mouse epidermis elevates apoptosis in BSCs. In contrast, ectopic Bcl-2 expression blocks apoptosis during HF regression, resulting in the accumulation of quiescent SCs and delaying HF growth in mice. Strikingly, Bcl-2-induced changes in size and composition of the HF bulge accelerate tumour formation. Our study identifies a niche-instructive mechanism of Bcl-2-regulated apoptosis response that is required for SC homeostasis and tissue regeneration, and may suppress carcinogenesis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Folículo Piloso , Animales , Apoptosis/genética , Ratones , Piel , Células Madre
4.
Br J Cancer ; 120(1): 69-78, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30377339

RESUMEN

BACKGROUND: Immune infiltration is implicated in the development of acquired resistance to anti-angiogenic cancer therapy. We therefore investigated the correlation between neutrophil infiltration in metastasis of colorectal cancer (CRC) patients and survival after treatment with bevacizumab. Our study identifies CD177+ tumour neutrophil infiltration as an adverse prognostic factor for bevacizumab treatment. We further demonstrate that a novel anti-VEGF/anti-Ang2 compound (BI-880) can overcome resistance to VEGF inhibition in experimental tumour models. METHODS: A total of 85 metastatic CRC patients were stratified into cohorts that had either received chemotherapy alone (n = 39) or combined with bevacizumab (n = 46). Tumour CD177+ neutrophil infiltration was correlated to clinical outcome. The impact of neutrophil infiltration on anti-VEGF or anti-VEGF/anti-Ang2 therapy was studied in both xenograft and syngeneic tumour models by immunohistochemistry. RESULTS: The survival of bevacizumab-treated CRC patients in the presence of CD177+ infiltrates was significantly reduced compared to patients harbouring CD177- metastases. BI-880 treatment reduced the development of hypoxia associated with bevacizumab treatment and improved vascular normalisation in xenografts. Furthermore, neutrophil depletion or BI-880 treatment restored treatment sensitivity in a syngeneic tumour model of anti-VEGF resistance. CONCLUSIONS: Our findings implicate CD177 as a biomarker for bevacizumab and suggest VEGF/Ang2 inhibition as a strategy to overcome neutrophil associated resistance to anti-angiogenic treatment.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Isoantígenos/genética , Neovascularización Patológica/tratamiento farmacológico , Receptores de Superficie Celular/genética , Factor A de Crecimiento Endotelial Vascular/genética , Proteínas de Transporte Vesicular/genética , Anciano , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Bevacizumab/administración & dosificación , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos/genética , Femenino , Proteínas Ligadas a GPI/genética , Humanos , Masculino , Ratones , Persona de Mediana Edad , Metástasis de la Neoplasia , Células Madre Neoplásicas/efectos de los fármacos , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Neutrófilos/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
5.
EMBO J ; 33(19): 2171-87, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25056906

RESUMEN

The X-linked inhibitor of apoptosis protein (XIAP) is a potent caspase inhibitor, best known for its anti-apoptotic function in cancer. During apoptosis, XIAP is antagonized by SMAC, which is released from the mitochondria upon caspase-mediated activation of BID. Recent studies suggest that XIAP is involved in immune signaling. Here, we explore XIAP as an important mediator of an immune response against the enteroinvasive bacterium Shigella flexneri, both in vitro and in vivo. Our data demonstrate for the first time that Shigella evades the XIAP-mediated immune response by inducing the BID-dependent release of SMAC from the mitochondria. Unlike apoptotic stimuli, Shigella activates the calpain-dependent cleavage of BID to trigger the release of SMAC, which antagonizes the inflammatory action of XIAP without inducing apoptosis. Our results demonstrate how the cellular death machinery can be subverted by an invasive pathogen to ensure bacterial colonization.


Asunto(s)
Proteína Proapoptótica que Interacciona Mediante Dominios BH3/fisiología , Proteínas Portadoras/metabolismo , Disentería Bacilar/inmunología , Mitocondrias/inmunología , Proteínas Mitocondriales/metabolismo , Shigella/inmunología , Proteína Inhibidora de la Apoptosis Ligada a X/fisiología , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis , Western Blotting , Caspasas/metabolismo , Proliferación Celular , Células Cultivadas , Disentería Bacilar/microbiología , Disentería Bacilar/patología , Femenino , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/patología , Técnicas para Inmunoenzimas , Integrasas/metabolismo , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Proteínas Mitocondriales/inmunología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Shigella/patogenicidad , Transducción de Señal , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem
6.
Blood ; 127(22): 2732-41, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27048211

RESUMEN

The adaptor protein MYD88 is critical for relaying activation of Toll-like receptor signaling to NF-κB activation. MYD88 mutations, particularly the p.L265P mutation, have been described in numerous distinct B-cell malignancies, including diffuse large B-cell lymphoma (DLBCL). Twenty-nine percent of activated B-cell-type DLBCL (ABC-DLBCL), which is characterized by constitutive activation of the NF-κB pathway, carry the p.L265P mutation. In addition, ABC-DLBCL frequently displays focal copy number gains affecting BCL2 Here, we generated a novel mouse model in which Cre-mediated recombination, specifically in B cells, leads to the conditional expression of Myd88(p.L252P) (the orthologous position of the human MYD88(p.L265P) mutation) from the endogenous locus. These mice develop a lymphoproliferative disease and occasional transformation into clonal lymphomas. The clonal disease displays the morphologic and immunophenotypical characteristics of ABC-DLBCL. Lymphomagenesis can be accelerated by crossing in a further novel allele, which mediates conditional overexpression of BCL2 Cross-validation experiments in human DLBCL samples revealed that both MYD88 and CD79B mutations are substantially enriched in ABC-DLBCL compared with germinal center B-cell DLBCL. Furthermore, analyses of human DLBCL genome sequencing data confirmed that BCL2 amplifications frequently co-occurred with MYD88 mutations, further validating our approach. Finally, in silico experiments revealed that MYD88-mutant ABC-DLBCL cells in particular display an actionable addiction to BCL2. Altogether, we generated a novel autochthonous mouse model of ABC-DLBCL that could be used as a preclinical platform for the development and validation of novel therapeutic approaches for the treatment of ABC-DLBCL.


Asunto(s)
Linfocitos B/metabolismo , Transformación Celular Neoplásica/metabolismo , Linfoma de Células B Grandes Difuso/metabolismo , Mutación Missense , Factor 88 de Diferenciación Mieloide/biosíntesis , Neoplasias Experimentales/metabolismo , Animales , Linfocitos B/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Humanos , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Ratones , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/genética
7.
EMBO Rep ; 16(6): 719-27, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25825408

RESUMEN

In this study, we show for the first time that the therapeutic antagonization of inhibitor of apoptosis proteins (IAPs) inhibits B16 melanoma growth by disrupting tumor vasculature. Specifically, the treatment of mice bearing B16 melanoma with an IAP antagonist compound A (Comp A) inhibits tumor growth not by inducing direct cytotoxicity against B16 cells but rather by a hitherto unrecognized antiangiogenic activity against tumor vessels. Our detailed analysis showed that Comp A treatment induces NF-κB activity in B16 tumor cells and facilitates the production of TNF. In the presence of Comp A, endothelial cells (ECs) become highly susceptible to TNF and undergo apoptotic cell death. Accordingly, the antiangiogenic and growth-attenuating effects of Comp A treatment were completely abolished in TNF-R knockout mice. This novel targeting approach could be of clinical value in controlling pathological neoangiogenesis under inflammatory condition while sparing blood vessels under normal condition.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Endotelio Vascular/patología , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Inflamación/fisiopatología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Neovascularización Patológica , Receptores del Factor de Necrosis Tumoral/genética , Factor de Necrosis Tumoral alfa/metabolismo
8.
Exp Dermatol ; 22(11): 719-24, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24112621

RESUMEN

Mast cells modulate autoimmune diseases such as psoriasis and multiple sclerosis. Fumaric acid esters (FAEs) are widely used for the treatment of psoriasis, and dimethylfumarate (DMF) has recently been approved for multiple sclerosis. In this study, we analysed the cytotoxic effect of FAEs on human mast cells. Specifically, cell death was analysed in the human mast cell line HMC-1 and in primary cord blood-derived mast cells (CBMCs) after incubation with fumaric acid (FA), monomethylfumarate (MMF), DMF and calcium bis(monomethylfumarate) (Ca-MF). Our data show that only DMF potently induces apoptotic cell death in HMC-1 cells and CBMCs. DMF-mediated apoptosis was associated with increased expression of Bax and Bak and activation of caspase-9 and caspase-6. Interestingly, DMF also enhanced the sensitivity of CBMCs towards TRAIL- and dexamethasone-induced apoptosis. These findings demonstrate for the first time that DMF induces apoptosis of human mast cells, primarily via the mitochondrial apoptotic pathway. Our study contributes to the understanding of the beneficial effects of FAEs in autoimmune diseases and provides a rationale for exploiting FAEs for other diseases associated with mast cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Fármacos Dermatológicos/química , Fumaratos/química , Mastocitos/efectos de los fármacos , Psoriasis/patología , Calcio/química , Caspasa 6/metabolismo , Caspasa 9/metabolismo , Muerte Celular , Línea Celular , Dexametasona/química , Dimetilfumarato , Etopósido/química , Humanos , Interleucina-8/metabolismo , Maleatos/química , Metotrexato/química , Ligando Inductor de Apoptosis Relacionado con TNF/química , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
9.
Cell Rep ; 38(7): 110370, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35172139

RESUMEN

The transition between quiescence and activation in neural stem and progenitor cells (NSPCs) is coupled with reversible changes in energy metabolism with key implications for lifelong NSPC self-renewal and neurogenesis. How this metabolic plasticity is ensured between NSPC activity states is unclear. We find that a state-specific rewiring of the mitochondrial proteome by the i-AAA peptidase YME1L is required to preserve NSPC self-renewal. YME1L controls the abundance of numerous mitochondrial substrates in quiescent NSPCs, and its deletion activates a differentiation program characterized by broad metabolic changes causing the irreversible shift away from a fatty-acid-oxidation-dependent state. Conditional Yme1l deletion in adult NSPCs in vivo results in defective self-renewal and premature differentiation, ultimately leading to NSPC pool depletion. Our results disclose an important role for YME1L in coordinating the switch between metabolic states of NSPCs and suggest that NSPC fate is regulated by compartmentalized changes in protein network dynamics.


Asunto(s)
Células Madre Adultas/metabolismo , Autorrenovación de las Células , Metaloendopeptidasas/metabolismo , Mitocondrias/enzimología , Células-Madre Neurales/metabolismo , Células Madre Adultas/citología , Animales , Proliferación Celular , Ciclo del Ácido Cítrico , Ácidos Grasos/metabolismo , Eliminación de Gen , Metaloendopeptidasas/deficiencia , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/ultraestructura , Células-Madre Neurales/citología , Nucleótidos/metabolismo , Oxidación-Reducción , Proteolisis , Proteoma/metabolismo
10.
PLoS Pathog ; 5(9): e1000596, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19779562

RESUMEN

Zwitterionic capsular polysaccharides (ZPS) of commensal bacteria are characterized by having both positive and negative charged substituents on each repeating unit of a highly repetitive structure that has an alpha-helix configuration. In this paper we look at the immune response of CD8(+) T cells to ZPSs. Intraperitoneal application of the ZPS Sp1 from Streptococcus pneumoniae serotype 1 induces CD8(+)CD28(-) T cells in the spleen and peritoneal cavity of WT mice. However, chemically modified Sp1 (mSp1) without the positive charge and resembling common negatively charged polysaccharides fails to induce CD8(+)CD28(-) T lymphocytes. The Sp1-induced CD8(+)CD28(-) T lymphocytes are CD122(low)CTLA-4(+)CD39(+). They synthesize IL-10 and TGF-beta. The Sp1-induced CD8(+)CD28(-) T cells exhibit immunosuppressive properties on CD4(+) T cells in vivo and in vitro. Experimental approaches to elucidate the mechanism of CD8(+) T cell activation by Sp1 demonstrate in a dimeric MHC class I-Ig model that Sp1 induces CD8(+) T cell activation by enhancing crosslinking of TCR. The expansion of CD8(+)CD28(-) T cells is independent, of direct antigen-presenting cell/T cell contact and, to the specificity of the T cell receptor (TCR). In CD8(+)CD28(-) T cells, Sp1 enhances Zap-70 phosphorylation and increasingly involves NF-kappaB which ultimately results in protection versus apoptosis and cell death and promotes survival and accumulation of the CD8(+)CD28(-) population. This is the first description of a naturally occurring bacterial antigen that is able to induce suppressive CD8(+)CD28(-) T lymphocytes in vivo and in vitro. The underlying mechanism of CD8(+) T cell activation appears to rely on enhanced TCR crosslinking. The data provides evidence that ZPS of commensal bacteria play an important role in peripheral tolerance mechanisms and the maintenance of the homeostasis of the immune system.


Asunto(s)
Antígenos Bacterianos/inmunología , Cápsulas Bacterianas/inmunología , Antígenos CD28/inmunología , Linfocitos T CD8-positivos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Streptococcus pneumoniae/inmunología , Linfocitos T Reguladores/inmunología , Absceso Abdominal/microbiología , Absceso Abdominal/patología , Animales , Células Presentadoras de Antígenos/inmunología , Apoptosis/inmunología , Citocinas/inmunología , Citometría de Flujo , Inmunohistoquímica , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
11.
Sci Rep ; 10(1): 17339, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33060693

RESUMEN

Mucosal and skin cancers are associated with infections by human papillomaviruses (HPV). The manner how viral oncoproteins hijack the host cell metabolism to meet their own energy demands and how this may contribute to tumorigenesis is poorly understood. We now show that the HPV oncoprotein E7 of HPV8, HPV11 and HPV16 directly interact with the beta subunit of the mitochondrial ATP-synthase (ATP5B), which may therefore represent a conserved feature across different HPV genera. By measuring both glycolytic and mitochondrial activity we observed that the association of E7 with ATP5B was accompanied by reduction of glycolytic activity. Interestingly, there was a drastic increase in spare mitochondrial respiratory capacity in HPV8-E7 and an even more profound increase in HPV16-E7 expressing cells. In addition, we could show that ATP5B levels were unchanged in betaHPV positive skin cancers. However, comparing HPV-positive and HPV-negative oropharyngeal squamous cell carcinomas (OPSCC) we noticed that, while ATP5B expression levels did not correlate with patient overall survival in HPV-negative OPSCC, there was a strong correlation within the HPV16-positive OPSCC patient group. These novel findings provide evidence that HPV targets the host cell energy metabolism important for viral life cycle and HPV-mediated tumorigenesis.


Asunto(s)
Alphapapillomavirus/aislamiento & purificación , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Neoplasias Orofaríngeas/virología , Infecciones por Papillomavirus/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/virología , Infecciones Tumorales por Virus/metabolismo , Femenino , Humanos , Proteínas Oncogénicas Virales/metabolismo , Fosforilación Oxidativa , Unión Proteica , Análisis de Supervivencia
12.
Nat Microbiol ; 5(2): 354-367, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31873204

RESUMEN

The cytosolic appearance and propagation of bacteria cause overwhelming cellular stress responses that induce apoptosis under normal conditions. Therefore, successful bacterial colonization depends on the ability of intracellular pathogens to block apoptosis and to safeguard bacterial replicative niches. Here, we show that the cytosolic Gram-negative bacterium Shigella flexneri stalls apoptosis by inhibiting effector caspase activity. Our data identified lipopolysaccharide (LPS) as a bona fide effector caspase inhibitor that directly binds caspases by involving its O-antigen (O Ag) moiety. Bacterial strains that lacked the O Ag or failed to replicate within the cytosol were incapable of blocking apoptosis and exhibited reduced virulence in a murine model of bacterial infection. Our findings demonstrate how Shigella inhibits pro-apoptotic caspase activity, effectively delays coordinated host-cell demise and supports its intracellular propagation. Next to the recently discovered pro-inflammatory role of cytosolic LPS, our data reveal a distinct mode of LPS action that, through the disruption of the early coordinated non-lytic cell death response, ultimately supports the inflammatory breakdown of infected cells at later time points.


Asunto(s)
Apoptosis/fisiología , Inhibidores de Caspasas/metabolismo , Caspasas Efectoras/metabolismo , Bacterias Gramnegativas/patogenicidad , Lipopolisacáridos/metabolismo , Shigella flexneri/patogenicidad , Animales , Citosol/microbiología , Femenino , Bacterias Gramnegativas/genética , Bacterias Gramnegativas/fisiología , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Antígenos O/metabolismo , Shigella flexneri/genética , Shigella flexneri/fisiología , Virulencia
13.
J Cell Biol ; 218(6): 1853-1870, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31085560

RESUMEN

In childhood, skeletal growth is driven by transient expansion of cartilage in the growth plate. The common belief is that energy production in this hypoxic tissue mainly relies on anaerobic glycolysis and not on mitochondrial respiratory chain (RC) activity. However, children with mitochondrial diseases causing RC dysfunction often present with short stature, which indicates that RC activity may be essential for cartilage-mediated skeletal growth. To elucidate the role of the mitochondrial RC in cartilage growth and pathology, we generated mice with impaired RC function in cartilage. These mice develop normally until birth, but their later growth is retarded. A detailed molecular analysis revealed that metabolic signaling and extracellular matrix formation is disturbed and induces cell death at the cartilage-bone junction to cause a chondrodysplasia-like phenotype. Hence, the results demonstrate the overall importance of the metabolic switch from fetal glycolysis to postnatal RC activation in growth plate cartilage and explain why RC dysfunction can cause short stature in children with mitochondrial diseases.


Asunto(s)
Cartílago/patología , Condrocitos/patología , Proteínas del Complejo de Cadena de Transporte de Electrón/antagonistas & inhibidores , Trastornos del Crecimiento/complicaciones , Placa de Crecimiento/patología , Enfermedades Mitocondriales/etiología , Animales , Cartílago/metabolismo , Diferenciación Celular , Condrocitos/metabolismo , Colágeno Tipo II/fisiología , ADN Helicasas/fisiología , Transporte de Electrón , Metabolismo Energético , Trastornos del Crecimiento/metabolismo , Trastornos del Crecimiento/patología , Placa de Crecimiento/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Proteínas Mitocondriales/fisiología , Transducción de Señal
14.
Cell Rep ; 25(9): 2354-2368.e5, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30485805

RESUMEN

IL-1ß is a cytokine of pivotal importance to the orchestration of inflammatory responses. Synthesized as an inactive pro-cytokine, IL-1ß requires proteolytic maturation to gain biological activity. Here, we identify intrinsic apoptosis as a non-canonical trigger of IL-1ß maturation. Guided by the discovery of the immunomodulatory activity of vioprolides, cyclic peptides isolated from myxobacteria, we observe IL-1ß maturation independent of canonical inflammasome pathways, yet dependent on intrinsic apoptosis. Mechanistically, vioprolides inhibit MCL-1 and BCL2, which in turn triggers BAX/BAK-dependent mitochondrial outer membrane permeabilization (MOMP). Induction of MOMP results in the release of pro-apoptotic factors initiating intrinsic apoptosis, as well as the depletion of IAPs (inhibitors of apoptosis proteins). IAP depletion, in turn, operates upstream of ripoptosome complex formation, subsequently resulting in caspase-8-dependent IL-1ß maturation. These results establish the ripoptosome/caspase-8 complex as a pro-inflammatory checkpoint that senses the perturbation of mitochondrial integrity.


Asunto(s)
Apoptosis , Caspasa 8/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , Caspasa 1/metabolismo , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , L-Lactato Deshidrogenasa/metabolismo , Ratones , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Péptidos Cíclicos/farmacología , Permeabilidad , Inhibidores de la Síntesis de la Proteína/farmacología
15.
Leuk Lymphoma ; 55(3): 645-51, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23697877

RESUMEN

Resistance to apoptosis is a hallmark of cancer, and represents an important mechanism of how tumor cells resist immune cell destruction. Mitochondria are the central regulators of the apoptotic machinery by releasing pro-apoptotic factors including cytochrome c and second mitochondria-derived activator of caspase (SMAC) upon mitochondrial outer membrane permeabilization (MOMP). Small molecules activating MOMP such as BH3 mimetics or antagonizers of the inhibitor of apoptosis proteins (IAPs) such as SMAC mimetics have recently engendered new optimism for a more individualized and effective cancer therapy. Here we show that a SMAC mimetic potentiates cancer cell killing by natural killer (NK) cells through reactivation of tumor cell apoptosis. Specifically, the SMAC mimetic enhances the susceptibility of tumor cells toward NK cell-mediated effector mechanisms involving death receptors and cytolytic granules containing perforin and granzymes by relieving caspase activity. Our data highlight for the first time the specific use of SMAC mimetics for boosting immune cell-mediated immunotherapy, representing a novel and promising approach in the treatment of cancer.


Asunto(s)
Citotoxicidad Inmunológica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Proteínas Mitocondriales/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Granzimas/metabolismo , Humanos , Mitocondrias/metabolismo , Perforina/metabolismo , Receptores de Muerte Celular/metabolismo
16.
EMBO Mol Med ; 6(5): 624-39, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24648500

RESUMEN

In the normal quiescent vasculature, only 0.01% of endothelial cells (ECs) are proliferating. However, this proportion increases dramatically following the angiogenic switch during tumor growth or wound healing. Recent evidence suggests that this angiogenic switch is accompanied by a metabolic switch. Here, we show that proliferating ECs increasingly depend on mitochondrial oxidative phosphorylation (OxPhos) for their increased energy demand. Under growth conditions, ECs consume three times more oxygen than quiescent ECs and work close to their respiratory limit. The increased utilization of the proton motif force leads to a reduced mitochondrial membrane potential in proliferating ECs and sensitizes to mitochondrial uncoupling. The benzoquinone embelin is a weak mitochondrial uncoupler that prevents neoangiogenesis during tumor growth and wound healing by exhausting the low respiratory reserve of proliferating ECs without adversely affecting quiescent ECs. We demonstrate that this can be exploited therapeutically by attenuating tumor growth in syngenic and xenograft mouse models. This novel metabolic targeting approach might be clinically valuable in controlling pathological neoangiogenesis while sparing normal vasculature and complementing cytostatic drugs in cancer treatment.


Asunto(s)
Benzoquinonas/farmacología , Respiración de la Célula/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Neovascularización Patológica , Neovascularización Fisiológica/efectos de los fármacos , Desacopladores/farmacología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Modelos Animales de Enfermedad , Ratones , Mitocondrias/metabolismo , Neoplasias/tratamiento farmacológico , Desacopladores/uso terapéutico , Cicatrización de Heridas/efectos de los fármacos
17.
Cell Rep ; 4(4): 669-80, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23954788

RESUMEN

Obesity increases the incidence of hepatocellular carcinoma (HCC) development in part through the activation of obesity-associated proinflammatory signaling. Here, we show that in lean mice, abrogation of IL-6Rα signaling protects against diethylnitrosamine (DEN)-induced HCC development. HCC protection occurs via Mcl-1 destabilization, thus promoting hepatocyte apoptosis. IL-6 regulates Mcl-1 stability via the inhibition of PP-1α expression, promoting GSK-3ß inactivation. In addition, IL-6 suppresses expression of the Mcl-1 E3 ligase (Mule). Consequently, IL-6Rα deficiency activates PP-1α and Mule expression, resulting in increased Mcl-1 turnover and protection against HCC development. In contrast, in obesity, inhibition of PP-1α and Mule expression, leading to Mcl-1 stabilization, occurs independently of IL-6 signaling. Collectively, this study provides evidence that obesity inhibits hepatocyte apoptosis through Mcl-1 stabilization independent of IL-6 signaling, thus promoting liver carcinogenesis.


Asunto(s)
Carcinogénesis/metabolismo , Neoplasias Hepáticas Experimentales/etiología , Neoplasias Hepáticas Experimentales/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Obesidad/complicaciones , Receptores de Interleucina-6/metabolismo , Animales , Apoptosis , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Neoplasias Hepáticas Experimentales/patología , Ratones , Ratones Noqueados , Obesidad/metabolismo , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Estabilidad Proteica , Receptores de Interleucina-6/genética , Transducción de Señal , Proteínas Supresoras de Tumor , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
18.
Cell Rep ; 3(3): 881-91, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23499448

RESUMEN

The BH3-only protein NOXA represents one of the critical mediators of DNA-damage-induced cell death. In particular, its involvement in cellular responses to cancer chemotherapy is increasingly evident. Here, we identify a strategy of cancer cells to escape genotoxic chemotherapy by increasing proteasomal degradation of NOXA. We show that the deubiquitylating enzyme UCH-L1 is a key regulator of NOXA turnover, which protects NOXA from proteasomal degradation by removing Lys(48)-linked polyubiquitin chains. In the majority of tumors from patients with melanoma or colorectal cancer suffering from high rates of chemoresistance, NOXA fails to accumulate because UCH-L1 expression is epigenetically silenced. Whereas UCH-L1/NOXA-positive tumor samples exhibit increased sensitivity to genotoxic chemotherapy, downregulation of UCH-L1 or inhibition of its deubiquitylase activity resulted in reduced NOXA stability and resistance to genotoxic chemotherapy in both human and C. elegans cells. Our data identify the UCH-L1/NOXA interaction as a therapeutic target for overcoming cancer chemoresistance.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Ubiquitinación , Animales , Antineoplásicos/uso terapéutico , Apoptosis , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Daño del ADN , Regulación hacia Abajo , Silenciador del Gen , Humanos , Melanoma/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Proteolisis , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Interferente Pequeño , Ubiquitina/metabolismo , Ubiquitina Tiolesterasa/genética
19.
Leuk Lymphoma ; 54(4): 835-42, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22989017

RESUMEN

Basophilia is associated with allergic and parasitic diseases and advanced chronic myeloid leukemia. In the present study, we characterized the expression and function of the death receptors Fas/CD95 and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors in basophils from healthy donors compared to neoplastic basophils. Peripheral blood basophils obtained from healthy donors (HD-PBB) and from patients with chronic myeloid leukemia (CML-PBB) were found to express high levels of Fas/CD95 and low levels of TRAIL-R2, whereas the basophil-like chronic myeloid leukemia cell line KU-812 expressed significant levels of TRAIL-R1 and TRAIL-R2. HD-PBB underwent apoptosis in response to anti-Fas/CD95, but showed resistance to TRAIL, unless they were co-treated with actinomycin D. Interestingly, CML-PBB and KU-812 cells exhibited the opposite response pattern with resistance to anti-Fas/CD95, but significant susceptibility to TRAIL-induced apoptosis. Our data show that anti-Fas/CD95 and TRAIL differentially regulate apoptosis of normal and neoplastic human basophils, which may direct the development of novel therapeutic strategies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Apoptosis/efectos de los fármacos , Basófilos/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Receptor fas/antagonistas & inhibidores , Basófilos/metabolismo , Basófilos/patología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ciclohexanos , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Receptor fas/metabolismo
20.
Innate Immun ; 16(5): 310-21, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19897529

RESUMEN

Abscess formation associated with secondary peritonitis causes severe morbidity and can be fatal. Formation of abscesses requires the presence of CD4+ T-cells. Zwitterionic polysaccharides (ZPSs) represent a novel class of immunomodulatory bacterial antigens that stimulate CD4+ T-cells in a major histocompatibility complex (MHC) class II-dependent manner. The capsular polysaccharide Sp1 of Streptococcus pneumoniae serotype 1 possesses a zwitterionic charge with free amino groups and promotes T-cell-dependent abscess formation in an experimental mouse model. So far, nothing is known about the function of Interleukin (IL)-6 in intraperitoneal abscess formation. Here, we demonstrate that macrophages and dendritic cells (DCs), the most prevalent professional antigen-presenting cells involved in the formation of abscesses, secrete Interleukin (IL)-6 and are incorporated in the abscess capsule. Sp1 inhibits apoptosis of CD4+ T-cells and causes IL-17 expression by CD4+ T-cells in an IL-6-dependent manner. Abrogation of the Sp1-induced pleiotropic effects of IL-6 in IL-6-deficient mice and mice treated with an IL-6-specific neutralizing antibody results in significant inhibition of abscess formation. The data delineate the essential role of IL-6 in the linkage of innate and adaptive immunity in polysaccharide-mediated abscess formation.


Asunto(s)
Absceso/inmunología , Células Presentadoras de Antígenos/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Células Dendríticas/metabolismo , Streptococcus pneumoniae/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/patología , Cápsulas Bacterianas/inmunología , Cápsulas Bacterianas/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Movimiento Celular , Supervivencia Celular , Células Dendríticas/inmunología , Células Dendríticas/patología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Cavidad Peritoneal/patología , Polisacáridos Bacterianos/química , Polisacáridos Bacterianos/inmunología , Polisacáridos Bacterianos/metabolismo , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA