Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 195
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunol Rev ; 321(1): 350-370, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38093416

RESUMEN

Dendritic cells (DCs) are myeloid cells bridging the innate and adaptive immune system. By cross-presenting tumor-associated antigens (TAAs) liberated upon spontaneous or therapy-induced tumor cell death to T cells, DCs occupy a pivotal position in the cancer immunity cycle. Over the last decades, the mechanisms linking cancer cell death to DC maturation, have been the focus of intense research. Growing evidence supports the concept that the mere transfer of TAAs during the process of cell death is insufficient to drive immunogenic DC maturation unless this process is coupled with the release of immunomodulatory signals by dying cancer cells. Malignant cells succumbing to a regulated cell death variant called immunogenic cell death (ICD), foster a proficient interface with DCs, enabling their immunogenic maturation and engagement of adaptive immunity against cancer. This property relies on the ability of ICD to exhibit pathogen-mimicry hallmarks and orchestrate the emission of a spectrum of constitutively present or de novo-induced danger signals, collectively known as damage-associated molecular patterns (DAMPs). In this review, we discuss how DCs perceive and decode danger signals emanating from malignant cells undergoing ICD and provide an outlook of the major signaling and functional consequences of this interaction for DCs and antitumor immunity.


Asunto(s)
Muerte Celular Inmunogénica , Neoplasias , Humanos , Células Dendríticas , Muerte Celular , Antígenos de Neoplasias , Inmunidad Adaptativa
2.
Nature ; 579(7797): 111-117, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32103177

RESUMEN

The avascular nature of cartilage makes it a unique tissue1-4, but whether and how the absence of nutrient supply regulates chondrogenesis remain unknown. Here we show that obstruction of vascular invasion during bone healing favours chondrogenic over osteogenic differentiation of skeletal progenitor cells. Unexpectedly, this process is driven by a decreased availability of extracellular lipids. When lipids are scarce, skeletal progenitors activate forkhead box O (FOXO) transcription factors, which bind to the Sox9 promoter and increase its expression. Besides initiating chondrogenesis, SOX9 acts as a regulator of cellular metabolism by suppressing oxidation of fatty acids, and thus adapts the cells to an avascular life. Our results define lipid scarcity as an important determinant of chondrogenic commitment, reveal a role for FOXO transcription factors during lipid starvation, and identify SOX9 as a critical metabolic mediator. These data highlight the importance of the nutritional microenvironment in the specification of skeletal cell fate.


Asunto(s)
Huesos/citología , Microambiente Celular , Condrogénesis , Metabolismo de los Lípidos , Factor de Transcripción SOX9/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Huesos/irrigación sanguínea , Condrocitos/citología , Condrocitos/metabolismo , Ácidos Grasos/metabolismo , Femenino , Privación de Alimentos , Factores de Transcripción Forkhead/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Osteogénesis , Oxidación-Reducción , Factor de Transcripción SOX9/genética , Transducción de Señal , Cicatrización de Heridas
3.
Nature ; 578(7795): 419-424, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31996848

RESUMEN

ATP13A2 (PARK9) is a late endolysosomal transporter that is genetically implicated in a spectrum of neurodegenerative disorders, including Kufor-Rakeb syndrome-a parkinsonism with dementia1-and early-onset Parkinson's disease2. ATP13A2 offers protection against genetic and environmental risk factors of Parkinson's disease, whereas loss of ATP13A2 compromises lysosomes3. However, the transport function of ATP13A2 in lysosomes remains unclear. Here we establish ATP13A2 as a lysosomal polyamine exporter that shows the highest affinity for spermine among the polyamines examined. Polyamines stimulate the activity of purified ATP13A2, whereas ATP13A2 mutants that are implicated in disease are functionally impaired to a degree that correlates with the disease phenotype. ATP13A2 promotes the cellular uptake of polyamines by endocytosis and transports them into the cytosol, highlighting a role for endolysosomes in the uptake of polyamines into cells. At high concentrations polyamines induce cell toxicity, which is exacerbated by ATP13A2 loss due to lysosomal dysfunction, lysosomal rupture and cathepsin B activation. This phenotype is recapitulated in neurons and nematodes with impaired expression of ATP13A2 or its orthologues. We present defective lysosomal polyamine export as a mechanism for lysosome-dependent cell death that may be implicated in neurodegeneration, and shed light on the molecular identity of the mammalian polyamine transport system.


Asunto(s)
Lisosomas/metabolismo , Poliaminas/metabolismo , ATPasas de Translocación de Protón/deficiencia , ATPasas de Translocación de Protón/genética , Animales , Biocatálisis , Transporte Biológico , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Catepsina B/metabolismo , Citosol/metabolismo , Modelos Animales de Enfermedad , Endocitosis , Humanos , Lisosomas/patología , Ratones , Mutación , Neuronas/metabolismo , Fenotipo , Poliaminas/toxicidad , ATPasas de Translocación de Protón/metabolismo , Espermidina/metabolismo , Espermina/metabolismo
4.
Trends Biochem Sci ; 46(12): 960-975, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34384657

RESUMEN

Intracellular iron fulfills crucial cellular processes, including DNA synthesis and mitochondrial metabolism, but also mediates ferroptosis, a regulated form of cell death driven by lipid-based reactive oxygen species (ROS). Beyond their established role in degradation and recycling, lysosomes occupy a central position in iron homeostasis and integrate metabolic and cell death signals emanating from different subcellular sites. We discuss the central role of the lysosome in preserving iron homeostasis and provide an integrated outlook of the regulatory circuits coupling the lysosomal system to the control of iron trafficking, interorganellar crosstalk, and ferroptosis induction. We also discuss novel studies unraveling how deregulated lysosomal iron-handling functions contribute to cancer, neurodegeneration, and viral infection, and can be harnessed for therapeutic interventions.


Asunto(s)
Ferroptosis , Muerte Celular/fisiología , Hierro/metabolismo , Lisosomas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
5.
EMBO J ; 40(10): e106214, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33932034

RESUMEN

BNIP3 is a mitophagy receptor with context-dependent roles in cancer, but whether and how it modulates melanoma growth in vivo remains unknown. Here, we found that elevated BNIP3 levels correlated with poorer melanoma patient's survival and depletion of BNIP3 in B16-F10 melanoma cells compromised tumor growth in vivo. BNIP3 depletion halted mitophagy and enforced a PHD2-mediated downregulation of HIF-1α and its glycolytic program both in vitro and in vivo. Mechanistically, we found that BNIP3-deprived melanoma cells displayed increased intracellular iron levels caused by heightened NCOA4-mediated ferritinophagy, which fostered PHD2-mediated HIF-1α destabilization. These effects were not phenocopied by ATG5 or NIX silencing. Restoring HIF-1α levels in BNIP3-depleted melanoma cells rescued their metabolic phenotype and tumor growth in vivo, but did not affect NCOA4 turnover, underscoring that these BNIP3 effects are not secondary to HIF-1α. These results unravel an unexpected role of BNIP3 as upstream regulator of the pro-tumorigenic HIF-1α glycolytic program in melanoma cells.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Melanoma/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Línea Celular Tumoral , Biología Computacional , Femenino , Cromatografía de Gases y Espectrometría de Masas , Humanos , Immunoblotting , Inmunohistoquímica , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/genética , Transducción de Señal/fisiología
6.
Mol Cell ; 65(5): 885-899.e6, 2017 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-28238652

RESUMEN

Loss of ER Ca2+ homeostasis triggers endoplasmic reticulum (ER) stress and drives ER-PM contact sites formation in order to refill ER-luminal Ca2+. Recent studies suggest that the ER stress sensor and mediator of the unfolded protein response (UPR) PERK regulates intracellular Ca2+ fluxes, but the mechanisms remain elusive. Here, using proximity-dependent biotin identification (BioID), we identified the actin-binding protein Filamin A (FLNA) as a key PERK interactor. Cells lacking PERK accumulate F-actin at the cell edges and display reduced ER-PM contacts. Following ER-Ca2+ store depletion, the PERK-FLNA interaction drives the expansion of ER-PM juxtapositions by regulating F-actin-assisted relocation of the ER-associated tethering proteins Stromal Interaction Molecule 1 (STIM1) and Extended Synaptotagmin-1 (E-Syt1) to the PM. Cytosolic Ca2+ elevation elicits rapid and UPR-independent PERK dimerization, which enforces PERK-FLNA-mediated ER-PM juxtapositions. Collectively, our data unravel an unprecedented role of PERK in the regulation of ER-PM appositions through the modulation of the actin cytoskeleton.


Asunto(s)
Citoesqueleto de Actina/enzimología , Actinas/metabolismo , Membrana Celular/enzimología , Estrés del Retículo Endoplásmico , Retículo Endoplásmico/enzimología , Filaminas/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio , Filaminas/genética , Células HEK293 , Células HeLa , Humanos , Ratones , Proteínas de Neoplasias/metabolismo , Multimerización de Proteína , Transporte de Proteínas , Interferencia de ARN , Transducción de Señal , Molécula de Interacción Estromal 1/metabolismo , Sinaptotagmina I/metabolismo , Factores de Tiempo , Transfección , Respuesta de Proteína Desplegada , eIF-2 Quinasa/genética
7.
Acta Neuropathol ; 147(1): 6, 2024 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-38170217

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a rapidly progressive and fatal neurodegenerative disorder, characterized by selective loss of motor neurons (MNs). A number of causative genetic mutations underlie the disease, including mutations in the fused in sarcoma (FUS) gene, which can lead to both juvenile and late-onset ALS. Although ALS results from MN death, there is evidence that dysfunctional glial cells, including oligodendroglia, contribute to neurodegeneration. Here, we used human induced pluripotent stem cells (hiPSCs) with a R521H or a P525L mutation in FUS and their isogenic controls to generate oligodendrocyte progenitor cells (OPCs) by inducing SOX10 expression from a TET-On SOX10 cassette. Mutant and control iPSCs differentiated efficiently into OPCs. RNA sequencing identified a myelin sheath-related phenotype in mutant OPCs. Lipidomic studies demonstrated defects in myelin-related lipids, with a reduction of glycerophospholipids in mutant OPCs. Interestingly, FUSR521H OPCs displayed a decrease in the phosphatidylcholine/phosphatidylethanolamine ratio, known to be associated with maintaining membrane integrity. A proximity ligation assay further indicated that mitochondria-associated endoplasmic reticulum membranes (MAM) were diminished in both mutant FUS OPCs. Moreover, both mutant FUS OPCs displayed increased susceptibility to ER stress when exposed to thapsigargin, and exhibited impaired mitochondrial respiration and reduced Ca2+ signaling from ER Ca2+ stores. Taken together, these results demonstrate a pathological role of mutant FUS in OPCs, causing defects in lipid metabolism associated with MAM disruption manifested by impaired mitochondrial metabolism with increased susceptibility to ER stress and with suppressed physiological Ca2+ signaling. As such, further exploration of the role of oligodendrocyte dysfunction in the demise of MNs is crucial and will provide new insights into the complex cellular mechanisms underlying ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Células Madre Pluripotentes Inducidas , Humanos , Esclerosis Amiotrófica Lateral/patología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas Motoras/metabolismo , Mutación , Oligodendroglía/metabolismo , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo
8.
Proc Natl Acad Sci U S A ; 117(49): 31198-31207, 2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33229544

RESUMEN

Recessive loss-of-function mutations in ATP13A2 (PARK9) are associated with a spectrum of neurodegenerative disorders, including Parkinson's disease (PD). We recently revealed that the late endo-lysosomal transporter ATP13A2 pumps polyamines like spermine into the cytosol, whereas ATP13A2 dysfunction causes lysosomal polyamine accumulation and rupture. Here, we investigate how ATP13A2 provides protection against mitochondrial toxins such as rotenone, an environmental PD risk factor. Rotenone promoted mitochondrial-generated superoxide (MitoROS), which was exacerbated by ATP13A2 deficiency in SH-SY5Y cells and patient-derived fibroblasts, disturbing mitochondrial functionality and inducing toxicity and cell death. Moreover, ATP13A2 knockdown induced an ATF4-CHOP-dependent stress response following rotenone exposure. MitoROS and ATF4-CHOP were blocked by MitoTEMPO, a mitochondrial antioxidant, suggesting that the impact of ATP13A2 on MitoROS may relate to the antioxidant properties of spermine. Pharmacological inhibition of intracellular polyamine synthesis with α-difluoromethylornithine (DFMO) also increased MitoROS and ATF4 when ATP13A2 was deficient. The polyamine transport activity of ATP13A2 was required for lowering rotenone/DFMO-induced MitoROS, whereas exogenous spermine quenched rotenone-induced MitoROS via ATP13A2. Interestingly, fluorescently labeled spermine uptake in the mitochondria dropped as a consequence of ATP13A2 transport deficiency. Our cellular observations were recapitulated in vivo, in a Caenorhabditis elegans strain deficient in the ATP13A2 ortholog catp-6 These animals exhibited a basal elevated MitoROS level, mitochondrial dysfunction, and enhanced stress response regulated by atfs-1, the C. elegans ortholog of ATF4, causing hypersensitivity to rotenone, which was reversible with MitoTEMPO. Together, our study reveals a conserved cell protective pathway that counters mitochondrial oxidative stress via ATP13A2-mediated lysosomal spermine export.


Asunto(s)
Factor de Transcripción Activador 4/genética , Adenosina Trifosfatasas/genética , Proteínas de Caenorhabditis elegans/genética , Mitocondrias/genética , ATPasas de Translocación de Protón/genética , Factores de Transcripción/genética , Animales , Caenorhabditis elegans , Eflornitina/farmacología , Fibroblastos/efectos de los fármacos , Lisosomas/genética , Lisosomas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Mutación/genética , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Poliaminas/metabolismo , Rotenona/farmacología , Espermina/metabolismo , Factor de Transcripción CHOP/genética
9.
Genes Immun ; 23(1): 1-11, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35046546

RESUMEN

Immunogenic cell death (ICD) has emerged as a key component of therapy-induced anti-tumor immunity. Over the past few years, ICD was found to play a pivotal role in a wide variety of novel and existing treatment modalities. The clinical application of these techniques in cancer treatment is still in its infancy. Glioblastoma (GBM) is the most lethal primary brain tumor with a dismal prognosis despite maximal therapy. The development of new therapies in this aggressive type of tumors remains highly challenging partially due to the cold tumor immune environment. GBM could therefore benefit from ICD-based therapies stimulating the anti-tumor immune response. In what follows, we will describe the mechanisms behind ICD and the ICD-based (pre)clinical advances in anticancer therapies focusing on GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Muerte Celular Inmunogénica , Pronóstico
10.
J Biol Chem ; 296: 100182, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33310703

RESUMEN

Polyamines, such as putrescine, spermidine, and spermine, are physiologically important polycations, but the transporters responsible for their uptake in mammalian cells remain poorly characterized. Here, we reveal a new component of the mammalian polyamine transport system using CHO-MG cells, a widely used model to study alternative polyamine uptake routes and characterize polyamine transport inhibitors for therapy. CHO-MG cells present polyamine uptake deficiency and resistance to a toxic polyamine biosynthesis inhibitor methylglyoxal bis-(guanylhydrazone) (MGBG), but the molecular defects responsible for these cellular characteristics remain unknown. By genome sequencing of CHO-MG cells, we identified mutations in an unexplored gene, ATP13A3, and found disturbed mRNA and protein expression. ATP13A3 encodes for an orphan P5B-ATPase (ATP13A3), a P-type transport ATPase that represents a candidate polyamine transporter. Interestingly, ATP13A3 complemented the putrescine transport deficiency and MGBG resistance of CHO-MG cells, whereas its knockdown in WT cells induced a CHO-MG phenotype demonstrated as a decrease in putrescine uptake and MGBG sensitivity. Taken together, our findings identify ATP13A3, which has been previously genetically linked with pulmonary arterial hypertension, as a major component of the mammalian polyamine transport system that confers sensitivity to MGBG.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Poliaminas/metabolismo , Putrescina/metabolismo , Adenosina Trifosfatasas/genética , Animales , Transporte Biológico , Células CHO , Cricetinae , Cricetulus , Inhibidores Enzimáticos/farmacología , Mitoguazona/farmacología , Mutación , Secuenciación Completa del Genoma/métodos
11.
Proc Natl Acad Sci U S A ; 116(1): 277-286, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30578322

RESUMEN

The mitochondrial intramembrane rhomboid protease PARL has been implicated in diverse functions in vitro, but its physiological role in vivo remains unclear. Here we show that Parl ablation in mouse causes a necrotizing encephalomyelopathy similar to Leigh syndrome, a mitochondrial disease characterized by disrupted energy production. Mice with conditional PARL deficiency in the nervous system, but not in muscle, develop a similar phenotype as germline Parl KOs, demonstrating the vital role of PARL in neurological homeostasis. Genetic modification of two major PARL substrates, PINK1 and PGAM5, do not modify this severe neurological phenotype. Parl-/- brain mitochondria are affected by progressive ultrastructural changes and by defects in Complex III (CIII) activity, coenzyme Q (CoQ) biosynthesis, and mitochondrial calcium metabolism. PARL is necessary for the stable expression of TTC19, which is required for CIII activity, and of COQ4, which is essential in CoQ biosynthesis. Thus, PARL plays a previously overlooked constitutive role in the maintenance of the respiratory chain in the nervous system, and its deficiency causes progressive mitochondrial dysfunction and structural abnormalities leading to neuronal necrosis and Leigh-like syndrome.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Enfermedad de Leigh/etiología , Metaloproteasas/deficiencia , Proteínas Mitocondriales/deficiencia , Ubiquinona/metabolismo , Animales , Encéfalo/metabolismo , Calcio/metabolismo , Enfermedad de Leigh/metabolismo , Enfermedad de Leigh/fisiopatología , Hígado/metabolismo , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Encefalomiopatías Mitocondriales/metabolismo , Encefalomiopatías Mitocondriales/fisiopatología , Músculo Esquelético/metabolismo , Especies Reactivas de Oxígeno/metabolismo
12.
Immunol Rev ; 280(1): 126-148, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29027218

RESUMEN

The immunogenicity of cancer cells is an emerging determinant of anti-cancer immunotherapy. Beyond developing immunostimulatory regimens like dendritic cell-based vaccines, immune-checkpoint blockers, and adoptive T-cell transfer, investigators are beginning to focus on the immunobiology of dying cancer cells and its relevance for the success of anticancer immunotherapies. It is currently accepted that cancer cells may die in response to anti-cancer therapies through regulated cell death programs, which may either repress or increase their immunogenic potential. In particular, the induction of immunogenic cancer cell death (ICD), which is hallmarked by the emission of damage-associated molecular patterns (DAMPs); molecules analogous to pathogen-associated molecular patterns (PAMPs) acting as danger signals/alarmins, is of great relevance in cancer therapy. These ICD-associated danger signals favor immunomodulatory responses that lead to tumor-associated antigens (TAAs)-directed T-cell immunity, which paves way for the removal of residual, treatment-resistant cancer cells. It is also emerging that cancer cells succumbing to ICD can orchestrate "altered-self mimicry" i.e. mimicry of pathogen defense responses, on the levels of nucleic acids and/or chemokines (resulting in type I interferon/IFN responses or pathogen response-like neutrophil activity). In this review, we exhaustively describe the main molecular, immunological, preclinical, and clinical aspects of immunosuppressive cell death or ICD (with respect to apoptosis, necrosis and necroptosis). We also provide an extensive historical background of these fields, with special attention to the self/non-self and danger models, which have shaped the field of cell death immunology.


Asunto(s)
Muerte Celular , Neoplasias/inmunología , Linfocitos T/inmunología , Alarminas/inmunología , Animales , Antígenos de Neoplasias/inmunología , Apoptosis , Autoantígenos/inmunología , Humanos , Inmunidad , Imitación Molecular , Piroptosis
13.
Nat Methods ; 14(3): 228-232, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28245209

RESUMEN

We argue that the field of extracellular vesicle (EV) biology needs more transparent reporting to facilitate interpretation and replication of experiments. To achieve this, we describe EV-TRACK, a crowdsourcing knowledgebase (http://evtrack.org) that centralizes EV biology and methodology with the goal of stimulating authors, reviewers, editors and funders to put experimental guidelines into practice.


Asunto(s)
Investigación Biomédica , Bases de Datos Bibliográficas , Vesículas Extracelulares/fisiología , Internacionalidad
14.
Trends Immunol ; 38(8): 577-593, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28610825

RESUMEN

Cancer immunotherapy is experiencing a renaissance spearheaded by immune checkpoint inhibitors (ICIs). This has spurred interest in 'upgrading' existing immunotherapies that previously experienced only sporadic success, such as dendritic cells (DCs) vaccines. In this review, we discuss the major molecular, immunological, and clinical determinants of existing first- and second-generation DC vaccines. We also outline the future trends for next-generation DC vaccines and describe their major hallmarks and prerequisites necessary for high anticancer efficacy. In addition, using existing data we compare DC vaccines with ICIs targeting CTLA4, PD1, and PD-L1, and argue that in various contexts next-generation DC vaccines are ready to meet some challenges currently confronting ICIs, thereby raising the need to integrate DC vaccines in future combinatorial immunotherapy regimens.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Células Dendríticas/inmunología , Inmunoterapia/tendencias , Neoplasias/terapia , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Quimioterapia Adyuvante , Humanos , Terapia Molecular Dirigida , Neoplasias/inmunología
15.
Apoptosis ; 24(3-4): 269-277, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30680482

RESUMEN

Smac mimetics that deplete cellular inhibitor of apoptosis (cIAP) proteins have been shown to activate Nuclear Factor-kappa B (NF-κB). Here, we report that Smac mimetic-mediated activation of NF-κB contributes to the rescue of cancer cells from tunicamycin (TM)-triggered apoptosis. The prototypic Smac mimetic BV6 activates non-canonical and canonical NF-κB pathways, while TM has little effect on NF-κB signaling. Importantly, ectopic expression of dominant-negative IκBα superrepressor (IκBα-SR), which inhibits canonical and non-canonical NF-κB activation, significantly reversed this BV6-imposed protection against TM. Similarly, transient or stable knockdown of NF-κB-inducing kinase, which accumulated upon exposure to BV6 alone and in combination with TM, significantly counteracted BV6-mediated inhibition of TM-induced apoptosis. Interestingly, while cIAP2 was initially degraded upon BV6 treatment, it was subsequently upregulated in an NF-κB-dependent manner, as this restoration of cIAP2 expression was abolished in IκBα-SR-overexpressing cells. Interestingly, upon exposure to TM/BV6 apoptosis was significantly increased in cIAP2 knockdown cells. Furthermore, NF-κB inhibition partially prevented BV6-stimulated expression of Mcl-1 upon TM treatment. Consistently, Mcl-1 silencing significantly inhibited BV6-mediated protection from TM-induced apoptosis. Thus, NF-κB activation by Smac mimetic contributes to Smac mimetic-mediated protection against TM-induced apoptosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , FN-kappa B/metabolismo , Tunicamicina/farmacología , Línea Celular , Células HEK293 , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
16.
Biol Chem ; 400(2): 187-193, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-29924728

RESUMEN

Aerobic glycolysis ('Warburg effect') is used by cancer cells to fuel tumor growth. Interestingly, metastatic melanoma cells rely on glutaminolysis rather than aerobic glycolysis for their bioenergetic needs through the tricarboxylic acid (TCA) cycle. Here, we compared the effects of glucose or glutamine on melanoma cell proliferation, migration and oxidative phosphorylation in vitro. We found that glutamine-driven melanoma cell's aggressive traits positively correlated with increased expression of HIF1α and its pro-autophagic target BNIP3. BNIP3 silencing reduced glutamine-mediated effects on melanoma cell growth, migration and bioenergetics. Hence, BNIP3 is a vital component of the mitochondria quality control required for glutamine-driven melanoma aggressiveness.


Asunto(s)
Glutamina/metabolismo , Melanoma/patología , Proteínas de la Membrana/fisiología , Proteínas Proto-Oncogénicas/fisiología , Neoplasias Cutáneas/patología , Línea Celular Tumoral , Proliferación Celular , Metabolismo Energético , Silenciador del Gen , Humanos , Melanoma/metabolismo , Proteínas de la Membrana/genética , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas/genética , Neoplasias Cutáneas/metabolismo
18.
Curr Top Microbiol Immunol ; 414: 73-102, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28349285

RESUMEN

The endoplasmic reticulum (ER) is a crucial organelle for coordinating cellular Ca2+ signaling and protein synthesis and folding. Moreover, the dynamic and complex membranous structures constituting the ER allow the formation of contact sites with other organelles and structures, including among others the mitochondria and the plasma membrane (PM). The contact sites that the ER form with mitochondria is a hot topic in research, and the nature of the so-called mitochondria-associated membranes (MAMs) is continuously evolving. The MAMs consist of a proteinaceous tether that physically connects the ER with mitochondria. The MAMs harness the main functions of both organelles to form a specialized subcompartment at the interface of the ER and mitochondria. Under homeostatic conditions, MAMs are crucial for the efficient transfer of Ca2+ from the ER to mitochondria, and for proper mitochondria bioenergetics and lipid synthesis. MAMs are also believed to be the master regulators of mitochondrial shape and motility, and to form a crucial site for autophagosome assembly. Not surprisingly, MAMs have been shown to be a hot spot for the transfer of stress signals from the ER to mitochondria, most notably under the conditions of loss of ER proteostasis, by engaging the unfolded protein response (UPR). In this chapter after an introduction on ER biology and ER stress, we will review the emerging and key signaling roles of the MAMs, which have a root in cellular processes and signaling cascades coordinated by the ER.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Membranas Mitocondriales/fisiología , Animales , Autofagosomas/fisiología , Señalización del Calcio/fisiología , Humanos , Pliegue de Proteína , Respuesta de Proteína Desplegada/fisiología
19.
Cancer Immunol Immunother ; 67(7): 1179-1180, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29737376

RESUMEN

This correction refers to our Short Communication published in Cancer Immunology Immunotherapy in the year 2012 [1]. It has come to our attention that some errors resulting from accidental oversight concerning incorrect deletion/replacement of temporary placeholder images during figure assembly and mounting occurred during the assembly of the "Intracellular Proteins" immunoblots presented in Fig. 1A and Fig. 1D.

20.
EMBO Rep ; 17(6): 800-10, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27113756

RESUMEN

The oxysterol-binding protein (OSBP)-related proteins ORP5 and ORP8 have been shown recently to transport phosphatidylserine (PS) from the endoplasmic reticulum (ER) to the plasma membrane (PM) at ER-PM contact sites. PS is also transferred from the ER to mitochondria where it acts as precursor for mitochondrial PE synthesis. Here, we show that, in addition to ER-PM contact sites, ORP5 and ORP8 are also localized to ER-mitochondria contacts and interact with the outer mitochondrial membrane protein PTPIP51. A functional lipid transfer (ORD) domain was required for this localization. Interestingly, ORP5 and ORP8 depletion leads to defects in mitochondria morphology and respiratory function.


Asunto(s)
Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Receptores de Esteroides/metabolismo , Línea Celular , Retículo Endoplásmico/ultraestructura , Técnicas de Silenciamiento del Gen , Humanos , Metabolismo de los Lípidos , Mitocondrias/genética , Mitocondrias/ultraestructura , Proteínas Mitocondriales/química , Proteínas Mitocondriales/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteínas Tirosina Fosfatasas/química , Proteínas Tirosina Fosfatasas/metabolismo , Receptores de Esteroides/química , Receptores de Esteroides/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA