Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Biol Int ; 45(7): 1448-1458, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33675282

RESUMEN

Ischemia/reperfusion (I/R) is a well-known injury to the myocardium, but the mechanism involved remains elusive. In addition to the well-accepted apoptosis theory, autophagy was recently found to be involved in the process, exerting a dual role as protection in ischemia and detriment in reperfusion. Activation of autophagy is mediated by mitochondrial permeability transition pore (MPTP) opening during reperfusion. In our previous study, we showed that MPTP opening is regulated by VDAC1, a channel protein located in the outer membrane of mitochondria. Thus, upregulation of VDAC1 expression is a possible trigger to cardiomyocyte autophagy via an unclear pathway. Here, we established an anoxia/reoxygenation (A/R) model in vitro to simulate the I/R process in vivo. At the end of A/R treatment, VDAC1, Beclin 1, and LC3-II/I were upregulated, and autophagic vacuoles were increased in cardiomyocytes, which showed a connection of VDAC1 and autophagy development. These variations also led to ROS burst, mitochondrial dysfunction, and aggravated apoptosis. Knockdown of VDAC1 by RNAi could alleviate the above-mentioned cellular damages. Additionally, the expression of PINK1 and Parkin was enhanced after A/R injury. Furthermore, Parkin was recruited to mitochondria from the cytosol, which suggested that the PINK1/Parkin autophagic pathway was activated during A/R. Nevertheless, the PINK1/Parkin pathway was effectively inhibited when VDAC1 was knocked-down. Taken together, the A/R-induced cardiomyocyte injury was mediated by VDAC1 upregulation, which led to cell autophagy via the PINK1/Parkin pathway, and finally aggravated apoptosis.


Asunto(s)
Mitocondrias/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Proteínas Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Animales , Apoptosis , Autofagia , Línea Celular , Potencial de la Membrana Mitocondrial , Miocitos Cardíacos , Ratas
2.
Cell Biol Int ; 44(11): 2178-2181, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32716117

RESUMEN

A recent study suggests that voltage-dependent anion channel (VDAC) oligomer pores promote mitochondrial outer membrane permeabilization and allow mitochondrial DNA (mtDNA) to be released into the cytosol in live cells. It challenges the notion that only occurs in apoptotic cells via BAX/BAK macropores. Cytosolic mtDNA activates cyclic GMP-AMP synthase (cGAS)-stimulator of IFN gene (STING) pathway and triggers type I interferon (IFN) response thereafter, which ultimately causes systemic lupus erythematosus. Mechanistically, mtDNA can interact with three positively charged residues (Lys12, Arg15, and Lys20) at the N-terminus of VDAC1, thereby strengthening VDAC1 oligomerization and facilitating mtDNA release. In addition, there are other pathways that can mediate mtDNA release, such as BAX/BAK macropores and virus-derived pores. The mtDNA released into the cytosol also triggers type I IFN response via the generally accepted cGAS-STING-TANK-binding kinase 1-IFN regulatory factor 3 axis. Collectively, VDAC oligomer pores provide us an attractive direction for us to understand mtDNA release-related diseases.


Asunto(s)
ADN Mitocondrial/metabolismo , Mitocondrias/metabolismo , Canales Aniónicos Dependientes del Voltaje/metabolismo , Apoptosis , Citosol/metabolismo , ADN Mitocondrial/genética , Membranas Mitocondriales/metabolismo , Transducción de Señal , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Canales Aniónicos Dependientes del Voltaje/fisiología , Proteína X Asociada a bcl-2/metabolismo
3.
Hum Mol Genet ; 26(13): 2493-2506, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28431142

RESUMEN

Despite considerable knowledge on the genetic basis of mitochondrial disorders, their pathophysiological consequences remain poorly understood. We previously used two-dimensional difference gel electrophoresis analyses to define a protein profile characteristic for respiratory chain complex III-deficiency that included a significant overexpression of cytosolic gelsolin (GSN), a cytoskeletal protein that regulates the severing and capping of the actin filaments. Biochemical and immunofluorescence assays confirmed a specific increase of GSN levels in the mitochondria from patients' fibroblasts and from transmitochondrial cybrids with complex III assembly defects. A similar effect was obtained in control cells upon treatment with antimycin A in a dose-dependent manner, showing that the enzymatic inhibition of complex III is sufficient to promote the mitochondrial localization of GSN. Mitochondrial subfractionation showed the localization of GSN to the mitochondrial outer membrane, where it interacts with the voltage-dependent anion channel protein 1 (VDAC1). In control cells, VDAC1 was present in five stable oligomeric complexes, which showed increased levels and a modified distribution pattern in the complex III-deficient cybrids. Downregulation of GSN expression induced cell death in both cell types, in parallel with the specific accumulation of VDAC1 dimers and the release of mitochondrial cytochrome c into the cytosol, indicating a role for GSN in the oligomerization of VDAC complexes and in the prevention of apoptosis. Our results demonstrate that respiratory chain complex III dysfunction induces the physiological upregulation and mitochondrial location of GSN, probably to promote cell survival responses through the modulation of the oligomeric state of the VDAC complexes.


Asunto(s)
Transporte de Electrón/fisiología , Gelsolina/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Antimicina A/metabolismo , Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular , Citocromos c/metabolismo , Fibroblastos/metabolismo , Gelsolina/genética , Células HeLa , Humanos , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Membranas Mitocondriales/metabolismo , Electroforesis Bidimensional Diferencial en Gel/métodos , Canal Aniónico 1 Dependiente del Voltaje/fisiología
4.
J Proteome Res ; 17(1): 698-709, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29185755

RESUMEN

Progesterone is a biphasic hormone whose confounding role in breast cancer cells involves an initial proliferative surge, followed by sustained growth arrest. Recently we reported that progesterone induces a time- and concentration-dependent release of reactive oxygen species and thus regulates the antiproliferative activity in the breast cancer cell line. Furthermore, the expression of p27, a crucial cell cycle control protein, was regulated by binding of progesterone on progesterone receptor B, thus leading to antiproliferative signaling via multiple signaling pathways including p53, PTEN, and antioxidant systems. Here, we performed an LC-MS/MS analysis of three different breast cancer cell lines. Bioinformatics data analysis and functional classification of proteins revealed a role of progesterone in calcium signaling in MCF-7 cells, and the major differentially expressed calcium regulators were S100A11, S100A10, calreticulin, VDAC1, SERCA3, and SERCA1. Later on we confirmed it by a cell-line-based system having a calcium cameleon sensor targeted at endoplasmic reticulum and found moderate calcium efflux from endoplasmic reticulum upon progesterone treatment. Real-time PCR, Western blot, and TMRM staining confirmed the role of calcium signaling regulators VDAC1 and SERCA3 in progesterone response. Taking together all of these results with our previous studies, we suggest that progesterone, by regulating important proteins involved in calcium signaling and transport, can modulate cell proliferation and cell death. Furthermore, our research may open new avenues for the hypothesis that surgery conducted during the luteal phase of the menstrual cycle might facilitate improved patient survival.


Asunto(s)
Neoplasias de la Mama/metabolismo , Señalización del Calcio/efectos de los fármacos , Progesterona/farmacología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/fisiología , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Neoplasias de la Mama/patología , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Células MCF-7 , Proteómica/métodos
5.
J Biol Chem ; 290(14): 9150-61, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-25681439

RESUMEN

Excessive Ca(2+) fluxes from the endoplasmic reticulum to the mitochondria result in apoptotic cell death. Bcl-2 and Bcl-XL proteins exert part of their anti-apoptotic function by directly targeting Ca(2+)-transport systems, like the endoplasmic reticulum-localized inositol 1,4,5-trisphosphate receptors (IP3Rs) and the voltage-dependent anion channel 1 (VDAC1) at the outer mitochondrial membranes. We previously demonstrated that the Bcl-2 homology 4 (BH4) domain of Bcl-2 protects against Ca(2+)-dependent apoptosis by binding and inhibiting IP3Rs, although the BH4 domain of Bcl-XL was protective independently of binding IP3Rs. Here, we report that in contrast to the BH4 domain of Bcl-2, the BH4 domain of Bcl-XL binds and inhibits VDAC1. In intact cells, delivery of the BH4-Bcl-XL peptide via electroporation limits agonist-induced mitochondrial Ca(2+) uptake and protects against staurosporine-induced apoptosis, in line with the results obtained with VDAC1(-/-) cells. Moreover, the delivery of the N-terminal domain of VDAC1 as a synthetic peptide (VDAC1-NP) abolishes the ability of BH4-Bcl-XL to suppress mitochondrial Ca(2+) uptake and to protect against apoptosis. Importantly, VDAC1-NP did not affect the ability of BH4-Bcl-2 to suppress agonist-induced Ca(2+) release in the cytosol or to prevent apoptosis, as done instead by an IP3R-derived peptide. In conclusion, our data indicate that the BH4 domain of Bcl-XL, but not that of Bcl-2, selectively targets VDAC1 and inhibits apoptosis by decreasing VDAC1-mediated Ca(2+) uptake into the mitochondria.


Asunto(s)
Apoptosis , Señalización del Calcio , Mitocondrias/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Proteína bcl-X/fisiología , Secuencia de Aminoácidos , Animales , Células Cultivadas , Ratones , Datos de Secuencia Molecular
6.
J Biol Chem ; 290(39): 23563-78, 2015 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-26253170

RESUMEN

The pro-apoptotic Bax and Bak proteins are considered central to apoptosis, yet apoptosis occurs in their absence. Here, we asked whether the mitochondrial protein VDAC1 mediates apoptosis independently of Bax/Bak. Upon screening a fungal secondary metabolite library for compounds inducing apoptosis in Bax/Bak-deficient mouse embryonic fibroblasts, we identified cyathin-R, a new cyathane diterpenoid compound able to activate apoptosis in the absence of Bax/Bak via promotion of the VDAC1 oligomerization that mediates cytochrome c release. Diphenylamine-2-carboxilic acid, an inhibitor of VDAC1 conductance and oligomerization, inhibited cyathin-R-induced VDAC1 oligomerization and apoptosis. Similarly, Bcl-2 overexpression conferred resistance to cyathin-R-induced apoptosis and VDAC1 oligomerization. Silencing of VDAC1 expression prevented cyathin-R-induced apoptosis. Finally, cyathin-R effectively attenuated tumor growth and induced apoptosis in Bax/Bak-deficient cells implanted into a xenograft mouse model. Hence, this study identified a new compound promoting VDAC1-dependent apoptosis as a potential therapeutic option for cancerous cells lacking or presenting inactivated Bax/Bak.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Diterpenos/farmacología , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína X Asociada a bcl-2/genética , Animales , Línea Celular Tumoral , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Hepáticas/efectos de los fármacos , Ratas
7.
Clin Lab ; 61(12): 1855-63, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26882807

RESUMEN

BACKGROUND: Although tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can selectively induce apoptosis in some non-small cell lung cancer (NSCLC) cells, some NSCLC cells exhibit TRAIL-resistance. The underlying mechanisms that regulate TRAIL sensitivity in NSCLC cells are not well understood. The objective of this study was to investigate molecular regulators of the TRAIL pathway in NSCLC cells. METHODS: The TRAIL-sensitive NSCLC cell line NCI-H358 and a TRAIL-resistant cell line A549 were treated with rmhTRAIL for 24 hours. Then cell viability were measured by MTT assay, meanwhile cell cycle and apoptosis were measured by flow cytometry. Furthermore, mass spectrometry (LC-MS/MS) was used to identify the difference in the protein expression profiles. Finally, real-time PCR was performed to detect the mRNA expression of TRAIL receptors and apoptotic related proteins. RESULTS: These results confirmed that NCI-H358 cells were sensitive to TRAIL, whereas A549 cells were resistant. Both mRNA and protein levels of voltage-dependent anion-selective channel proteinl (VDAC1), caspase9 (CASP9), and cytochrome c1 (CYC1) were upregulated in H358 cells but downregulated in A549 cells, whereas antiapoptotic protein BAG-2 was downregulated. In addition, TRAIL also causes DR5 low expression in A549 cells. CONCLUSIONS: These results indicate that rmhTRAIL had different anti-tumor activity in different NSCLC cell lines. Downregulation of VDAC1, CYC1, CASP9, and upregulation of BAG-2 might be associated with underlying TRAIL-resistance mechanisms. These findings motivated further studies to explore new therapeutic strategy overcoming TRAIL-resistance of NSCLC cells through modulating dysregulation of the proteins above.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/patología , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Espectrometría de Masas en Tándem , Canal Aniónico 1 Dependiente del Voltaje/fisiología
8.
Biochim Biophys Acta ; 1827(6): 793-805, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23541892

RESUMEN

Voltage-dependent anion selective channel isoform1 maintains the permeability of the outer mitochondrial membrane. Its voltage-gating properties are relevant in bioenergetic metabolism and apoptosis. The N-terminal domain is suspected to be involved in voltage-gating, due to its peculiar localization. However this issue is still controversial. In this work we exchanged or deleted the ß-strands that take contact with the N-terminal domain. The exchange of the whole hVDAC1 ß-barrel with the homologous hVDAC3 ß-barrel produces a chimeric protein that, in reconstituted systems, loses completely voltage-dependence. hVDAC3 ß-barrel has most residues in common with hVDAC1, including V143 and L150 considered anchor points for the N-terminus. hVDAC1 mutants completely lacking either the ß-strand 9 or both ß-strands 9 and 10 were expressed, refolded and reconstituted in artificial bilayers. The mutants formed smaller pores. Molecular dynamics simulations of the mutant structure supported its ability to form smaller pores. The mutant lacking both ß-strands 9 and 10 showed a new voltage-dependence feature resulting in a fully asymmetric behavior. These data indicate that a network of ß-strands in the pore-walls, and not single residues, are required for voltage-gating in addition to the N-terminus.


Asunto(s)
Canal Aniónico 1 Dependiente del Voltaje/química , Secuencia de Aminoácidos , Potenciales de la Membrana , Modelos Moleculares , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/química , Alineación de Secuencia , Canal Aniónico 1 Dependiente del Voltaje/fisiología
9.
Biochim Biophys Acta ; 1828(4): 1284-92, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23313453

RESUMEN

The voltage-dependent anion channel (VDAC) serves as the major pore for metabolites and electrolytes in the outer mitochondrial membrane. To refine our understanding of ion permeation through this channel we performed an extensive Brownian (BD) and molecular dynamics (MD) study on the mouse VDAC isoform 1 wild-type and mutants (K20E, D30K, K61E, E158K and K252E). The selectivity and the conductance of the wild-type and of the variant channels computed from the BD trajectories are in agreement with experimental data. The calculated selectivity is shown to be very sensitive to slight conformational changes which may have some bearing on the variability of the selectivity values measured on the VDAC open state. The MD and BD free energy profiles of the ion permeation suggest that the pore region comprising the N-terminal helix and the barrel band encircling it predominantly controls the ion transport across the channel. The overall 12µs BD and 0.9µs MD trajectories of the mouse VDAC isoform 1 wild-type and mutants feature no distinct pathways for ion diffusion and no long-lived ion-protein interactions. The dependence of ion distribution in the wild-type channel with the salt concentration can be explained by an ionic screening of the permanent charges of the protein arising from the pore. Altogether these results bolster the role of electrostatic features of the pore as the main determinant of VDAC selectivity towards inorganic anions.


Asunto(s)
Canales Aniónicos Dependientes del Voltaje/química , Canales Aniónicos Dependientes del Voltaje/fisiología , Animales , Conductividad Eléctrica , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Cloruro de Potasio/farmacología , Estructura Terciaria de Proteína , Electricidad Estática , Canal Aniónico 1 Dependiente del Voltaje/química , Canal Aniónico 1 Dependiente del Voltaje/fisiología
10.
Circulation ; 128(14): 1555-65, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23983249

RESUMEN

BACKGROUND: Under physiological conditions, Ca(2+) transfer from the endoplasmic reticulum (ER) to mitochondria might occur at least in part at contact points between the 2 organelles and involves the VDAC1/Grp75/IP3R1 complex. Accumulation of Ca(2+) into the mitochondrial matrix may activate the mitochondrial chaperone cyclophilin D (CypD) and trigger permeability transition pore opening, whose role in ischemia/reperfusion injury is well recognized. We questioned here whether the transfer of Ca(2+) from ER to mitochondria might play a role in cardiomyocyte death after hypoxia-reoxygenation. METHODS AND RESULTS: We report that CypD interacts with the VDAC1/Grp75/IP3R1 complex in cardiomyocytes. Genetic or pharmacological inhibition of CypD in both H9c2 cardiomyoblasts and adult cardiomyocytes decreased the Ca(2+) transfer from ER to mitochondria through IP3R under normoxic conditions. During hypoxia-reoxygenation, the interaction between CypD and the IP3R1 Ca(2+) channeling complex increased concomitantly with mitochondrial Ca(2+) content. Inhibition of either CypD, IP3R1, or Grp75 decreased protein interaction within the complex, attenuated mitochondrial Ca(2+) overload, and protected cells from hypoxia-reoxygenation. Genetic or pharmacological inhibition of CypD provided a similar effect in adult mice cardiomyocytes. Disruption of ER-mitochondria interaction via the downregulation of Mfn2 similarly reduced the interaction between CypD and the IP3R1 complex and protected against hypoxia-reoxygenation injury. CONCLUSIONS: Our data (1) point to a new role of CypD at the ER-mitochondria interface and (2) suggest that decreasing ER-mitochondria interaction at reperfusion can protect cardiomyocytes against lethal reperfusion injury through the reduction of mitochondrial Ca(2+) overload via the CypD/VDAC1/Grp75/IP3R1 complex.


Asunto(s)
Señalización del Calcio/fisiología , Hipoxia de la Célula/fisiología , Retículo Endoplásmico/fisiología , Mitocondrias Cardíacas/fisiología , Miocitos Cardíacos/patología , Oxígeno/toxicidad , Animales , Línea Celular , Células Cultivadas/metabolismo , Peptidil-Prolil Isomerasa F , Ciclofilinas/deficiencia , Ciclofilinas/genética , Ciclofilinas/fisiología , Proteínas HSP70 de Choque Térmico/fisiología , Técnicas In Vitro , Receptores de Inositol 1,4,5-Trifosfato/fisiología , Membranas Intracelulares/fisiología , Masculino , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Técnicas de Placa-Clamp , Distribución Aleatoria , Ratas , Canal Aniónico 1 Dependiente del Voltaje/fisiología
11.
Adv Exp Med Biol ; 772: 101-10, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24272356

RESUMEN

Finding new therapeutic targets to fight cancer is an ongoing quest. Because of insufficiencies in tumor vasculature, cells often are exposed to a hostile microenvironment that is low in oxygen (hypoxic) and nutrients. Thus, tumor cells face the challenge of finding new sources of energy and defying apoptosis, which allow them to survive, grow, and colonize other tissues. Eradicating specifically these hypoxic cells is one of the many goals of anticancer therapies. The mitochondrial voltage-dependent anion channel (VDAC) is a protein at the crossroads of metabolic and survival pathways. As its name suggests, VDAC is involved in ion transport as well as adenosine triphosphate and NAD(+) transport. We recently reported the presence in tumor cells of a novel hypoxia-induced form of VDAC. This form, a C-terminal truncated protein (VDAC1-ΔC), was associated in some cancer cell lines with a high output of adenosine triphosphate and a strong resistance to chemotherapy-induced apoptosis. Furthermore, VDAC1-ΔC was detected in tissues of 50 % of 46 patients with lung cancer. This review examines the significance of this new form of VDAC1 for anticancer therapy.


Asunto(s)
Biomarcadores de Tumor , Mitocondrias/genética , Neoplasias/terapia , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Animales , Apoptosis/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Hipoxia de la Célula/genética , Humanos , Mitocondrias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Isoformas de Proteínas/fisiología
12.
Proc Natl Acad Sci U S A ; 107(52): 22546-51, 2010 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-21148773

RESUMEN

The voltage-dependent anion channel (VDAC), located in the outer mitochondrial membrane, acts as a gatekeeper for the entry and exit of mitochondrial metabolites. Here we reveal functional dynamics of isoform one of VDAC (VDAC1) by a combination of solution NMR spectroscopy, Gaussian network model analysis, and molecular dynamics simulation. Micro- to millisecond dynamics are significantly increased for the N-terminal six ß-strands of VDAC1 in micellar solution, in agreement with increased B-factors observed in the same region in the bicellar crystal structure of VDAC1. Molecular dynamics simulations reveal that a charge on the membrane-facing glutamic acid 73 (E73) accounts for the elevation of N-terminal protein dynamics as well as a thinning of the nearby membrane. Mutation or chemical modification of E73 strongly reduces the micro- to millisecond dynamics in solution. Because E73 is necessary for hexokinase-I-induced VDAC channel closure and inhibition of apoptosis, our results imply that micro- to millisecond dynamics in the N-terminal part of the barrel are essential for VDAC interaction and gating.


Asunto(s)
Activación del Canal Iónico/fisiología , Espectroscopía de Resonancia Magnética/métodos , Simulación de Dinámica Molecular , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Animales , Cristalografía por Rayos X , Diciclohexilcarbodiimida/química , Dimiristoilfosfatidilcolina/química , Humanos , Cinética , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Ratones , Membranas Mitocondriales/química , Membranas Mitocondriales/metabolismo , Modelos Moleculares , Mutación Missense , Estructura Secundaria de Proteína , Soluciones , Factores de Tiempo , Canal Aniónico 1 Dependiente del Voltaje/química , Canal Aniónico 1 Dependiente del Voltaje/genética
13.
J Neurochem ; 119(5): 1137-50, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21951169

RESUMEN

Mitochondrial outer membrane Bax oligomers are critical for cytochrome c release, but the role of resident mitochondrial proteins in this process remains unclear. Membrane-associated Bax has primarily been studied using 3-[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonate (CHAPS) as the solubilizing agent, as it does not induce conformational artifacts, although recent evidence indicates it may have other artifactual effects. The objective of this study was to investigate digitonin as an alternative detergent to assess Bax oligomeric state, and possible interaction with voltage-dependent anion channel (VDAC)1 in cerebellar granule neurons. VDAC1 co-immunoprecipitated with Bax in digitonin extracts from healthy and apoptotic neurons. Two-dimensional blue native-SDS-PAGE revealed five Bax and VDAC1 oligomers having similar masses from 120 to 500 kDa. The levels of two VDAC1 oligomers in Bax 1D1 immunodepleted extracts negatively correlated with levels of co-precipitated VDAC1, indicating the co-precipitated VDAC1 was derived from these oligomers. Immunodepletion with the 6A7 antibody modestly reduced the levels of Bax oligomers from apoptotic but not healthy neurons. A sixth 170 kDa oligomer containing exclusively 6A7 Bax and no VDAC1 was identified after apoptosis induction. CHAPS failed to solubilize VDAC1, and additionally yielded no distinct oligomers. We conclude that digitonin is a potentially useful detergent preserving Bax-VDAC1 interactions that may be disrupted with CHAPS.


Asunto(s)
Corteza Cerebelosa/química , Gránulos Citoplasmáticos/química , Digitonina/farmacología , Neuronas/química , Canal Aniónico 1 Dependiente del Voltaje/química , Proteína X Asociada a bcl-2/química , Animales , Animales Recién Nacidos , Corteza Cerebelosa/citología , Corteza Cerebelosa/metabolismo , Ácidos Cólicos/farmacología , Gránulos Citoplasmáticos/metabolismo , Neuronas/clasificación , Neuronas/metabolismo , Cultivo Primario de Células , Ratas , Ratas Wistar , Solubilidad , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Proteína X Asociada a bcl-2/fisiología
14.
Am J Physiol Regul Integr Comp Physiol ; 300(6): R1311-5, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21389333

RESUMEN

The mitochondrial content of skeletal muscles is proportional to activity level, with the assumption that intrinsic mitochondrial function is the same in all muscles. This may not hold true for all muscles. For example, the diaphragm is a constantly active muscle; it is possible that its mitochondria are intrinsically different compared with other muscles. This study tested the hypothesis that mitochondrial respiration rates are greater in the diaphragm compared with triceps surae (TS, a limb muscle). We isolated mitochondria from diaphragm and TS of adult male Sprague Dawley rats. Mitochondrial respiration was measured by polarography. The contents of respiratory complexes, uncoupling proteins 1, 2, and 3 (UCP1, UCP2, and UCP3), and voltage-dependent anion channel 1 (VDAC1) were determined by immunoblotting. Complex IV activity was measured by spectrophotometry. Mitochondrial respiration states 3 (substrate and ADP driven) and 5 (uncoupled) were 27 ± 8% and 24 ± 10%, respectively, lower in diaphragm than in TS (P < 0.05 for both comparisons). However, the contents of respiratory complexes III, IV, and V, UCP1, and VDAC1 were higher in diaphragm mitochondria (23 ± 6, 30 ± 8, 25 ± 8, 36 ± 15, and 18 ± 8% respectively, P ≤ 0.04 for all comparisons). Complex IV activity was 64 ± 16% higher in diaphragm mitochondria (P ≤ 0.01). Mitochondrial UCP2 and UCP3 content and complex I activity were not different between TS and diaphragm. These data indicate that diaphragm mitochondria respire at lower rates, despite a higher content of respiratory complexes. The results invalidate our initial hypothesis and indicate that mitochondrial content is not the only determinant of aerobic capacity in the diaphragm. We propose that UCP1 and VDAC1 play a role in regulating diaphragm aerobic capacity.


Asunto(s)
Mitocondrias Musculares/fisiología , Músculo Esquelético/fisiología , Consumo de Oxígeno/fisiología , Músculos Respiratorios/fisiología , Animales , Respiración de la Célula/fisiología , Diafragma , Extremidades , Canales Iónicos/fisiología , Masculino , Proteínas Mitocondriales/fisiología , Modelos Animales , Ratas , Ratas Sprague-Dawley , Proteína Desacopladora 1 , Canal Aniónico 1 Dependiente del Voltaje/fisiología
15.
J Biol Chem ; 284(42): 28642-9, 2009 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-19717555

RESUMEN

Paraquat (PQ), a herbicide used worldwide, causes fatal injury to organs upon high dose ingestion. Treatments for PQ poisoning are unreliable, and numerous deaths have been attributed inappropriate usage of the agent. It is generally speculated that a microsomal drug-metabolizing enzyme system is responsible for PQ toxicity. However, recent studies have demonstrated cytotoxicity via mitochondria, and therefore, the cytotoxic mechanism remains controversial. Here, we demonstrated that mitochondrial NADH-dependent PQ reductase containing a voltage-dependent anion channel 1 (VDAC1) is responsible for PQ cytotoxicity. When mitochondria were incubated with NADH and PQ, superoxide anion (O(2)(*)) was produced, and the mitochondria ruptured. Outer membrane extract oxidized NADH in a PQ dose-dependent manner, and oxidation was suppressed by VDAC inhibitors. Zymographic analysis revealed the presence of VDAC1 protein in the oxidoreductase, and the direct binding of PQ to VDAC1 was demonstrated using biotinylated PQ. VDAC1-overexpressing cells showed increased O(2)(*) production and cytotoxicity, both of which were suppressed in VDAC1 knockdown cells. These results indicated that a VDAC1-containing mitochondrial system is involved in PQ poisoning. These insights into the mechanism of PQ poisoning not only demonstrated novel physiological functions of VDAC protein, but they may facilitate the development of new therapeutic approaches.


Asunto(s)
Herbicidas/toxicidad , NADH NADPH Oxidorreductasas/fisiología , NAD/metabolismo , Paraquat/toxicidad , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Animales , Sistema Libre de Células , Células HeLa , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Microscopía Electrónica/métodos , Mitocondrias/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno , Canal Aniónico 1 Dependiente del Voltaje/metabolismo
16.
Biochim Biophys Acta ; 1787(5): 421-30, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19094960

RESUMEN

The voltage-dependent anion channel (VDAC), located in the mitochondrial outer membrane, functions as gatekeeper for the entry and exit of mitochondrial metabolites, and thus controls cross-talk between mitochondria and the cytosol. VDAC also serves as a site for the docking of cytosolic proteins, such as hexokinase, and is recognized as a key protein in mitochondria-mediated apoptosis. The role of VDAC in apoptosis has emerged from various studies showing its involvement in cytochrome c release and apoptotic cell death as well as its interaction with proteins regulating apoptosis, including the mitochondria-bound isoforms of hexokinase (HK-I, HK-II). Recently, the functional HK-VDAC association has shifted from being considered in a predominantly metabolic light to the recognition of its major impact on the regulation of apoptotic responsiveness of the cell. Here, we demonstrate that the HK-VDAC1 interaction can be disrupted by mutating VDAC1 and by VDAC1-based peptides, consequently leading to diminished HK anti-apoptotic activity, suggesting that disruption of HK binding to VDAC1 can decrease tumor cell survival. Indeed, understanding structure-function relationships of VDAC is critical for deciphering how this channel can perform such a variety of differing functions, all important for cell life and death. By expressing VDAC1 mutants and VDAC1-based peptides, we have identified VDAC1 amino acid residues and domains important for interaction with HK and protection against apoptosis. These include negatively- and positively-charged residues, some of which are located within beta-strands of the protein. The N-terminal region of VDAC1 binds HK-I and prevents HK-mediated protection against apoptosis induced by STS, while expression of a VDAC N-terminal peptide detaches HK-I-GFP from mitochondria. These findings indicate that the interaction of HK with VDAC1 involves charged residues in several beta-strands and in the N-terminal domain. Displacing HK, serving as the 'guardian of the mitochondrion', from its binding site on VDAC1 may thus be exploited as an approach to cancer therapy.


Asunto(s)
Hexoquinasa/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Apoptosis , Neoplasias de la Mama , Línea Celular , Línea Celular Tumoral , Femenino , Células HeLa , Homeostasis , Humanos , Canales Iónicos/fisiología , Riñón , Cinética , Transfección , Células U937 , Canal Aniónico 1 Dependiente del Voltaje/química , Canal Aniónico 1 Dependiente del Voltaje/genética
17.
Theranostics ; 10(6): 2696-2713, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194829

RESUMEN

Rationale: Renal cell carcinoma (RCC) accounts for about 2% of all adult cancers, and clear cell RCC (ccRCC) is the most common RCC histologic subtype. A hallmark of ccRCC is the loss of the primary cilium, a cellular antenna that senses a wide variety of signals. Loss of this key organelle in ccRCC is associated with the loss of the von Hippel-Lindau protein (VHL). However, not all mechanisms of ciliopathy have been clearly elucidated. Methods: By using RCC4 renal cancer cells and patient samples, we examined the regulation of ciliogenesis via the presence or absence of the hypoxic form of the voltage-dependent anion channel (VDAC1-ΔC) and its impact on tumor aggressiveness. Three independent cohorts were analyzed. Cohort A was from PREDIR and included 12 patients with hereditary pVHL mutations and 22 sporadic patients presenting tumors with wild-type pVHL or mutated pVHL; Cohort B included tissue samples from 43 patients with non-metastatic ccRCC who had undergone surgery; and Cohort C was composed of 375 non-metastatic ccRCC tumor samples from The Cancer Genome Atlas (TCGA) and was used for validation. The presence of VDAC1-ΔC and legumain was determined by immunoblot. Transcriptional regulation of IFT20/GLI1 expression was evaluated by qPCR. Ciliogenesis was detected using both mouse anti-acetylated α-tubulin and rabbit polyclonal ARL13B antibodies for immunofluorescence. Results: Our study defines, for the first time, a group of ccRCC patients in which the hypoxia-cleaved form of VDAC1 (VDAC1-ΔC) induces resorption of the primary cilium in a Hypoxia-Inducible Factor-1 (HIF-1)-dependent manner. An additional novel group, in which the primary cilium is re-expressed or maintained, lacked VDAC1-ΔC yet maintained glycolysis, a signature of epithelial-mesenchymal transition (EMT) and more aggressive tumor progression, but was independent to VHL. Moreover, these patients were less sensitive to sunitinib, the first-line treatment for ccRCC, but were potentially suitable for immunotherapy, as indicated by the immunophenoscore and the presence of PDL1 expression. Conclusion: This study provides a new way to classify ccRCC patients and proposes potential therapeutic targets linked to metabolism and immunotherapy.


Asunto(s)
Carcinoma de Células Renales , Cilios , Neoplasias Renales , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Cilios/metabolismo , Cilios/patología , Estudios de Cohortes , Transición Epitelial-Mesenquimal , Femenino , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Adulto Joven
18.
Mol Genet Metab ; 97(4): 241-3, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19487148

RESUMEN

The primary structure of native human type-1 voltage dependent anion-selective channel/porin was presented twenty years ago, so was first data on its extra-mitochondrial expression in cell membranes of lymphocytes. Then, the channel had already entered cancer research as the docking molecule for hexokinase at outer mitochondrial membrane. Cell membrane standing porin met the cancer field only four years ago, when it was reported that normal and cancerous prostate cells from a single patient differed in the expression level of the channel. Meanwhile studies on a role of VDAC in cell differentiation, apoptosis, cancer and even pharmacology increase, mostly focused on porin in the outer membrane of mitochondria, but sometimes also pointing to the channel in the plasmalemma, e.g. prostate cancer cells on their way to neuroendocrine-differentiation. The synopsis presented discusses some recent papers on this issue, and argues in favor of considering voltage dependent anion-selective channel-cored volume regulated anion channel complexes in studies focused on apoptosis and cancer, where the channel might be part of the extrinsic cell death pathway. In this context, heed should also be given to the interaction of extra-mitochondrial porin and estrogen receptors alpha in cell membrane caveolae. Finally, it is insinuated to search for natural antibodies against type-1 porin, what in combination with other established markers might help in early diagnosis of prostate cancer.


Asunto(s)
Apoptosis/fisiología , Diferenciación Celular/efectos de los fármacos , Neoplasias de la Próstata/patología , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Línea Celular Tumoral , Humanos , Masculino
19.
FASEB J ; 22(8): 2809-20, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18381814

RESUMEN

Endostatin (ES) was reported to stimulate apoptosis in endothelial cells, but the exact mechanism remains controversial. In the present study, we elucidate the mechanism of ES-induced endothelial cell apoptosis. Our results indicate that ES induces cytochrome c release and caspase-9 activation in human microvascular endothelial cells (HMECs) at the concentration of 1 microM for 24 h, which initiates the apoptosis process. Further, ATP production, mitochondrial membrane potential, and tubule formation assays showed that ES promotes the mitochondrial permeability transition pore (mPTP) opening via voltage-dependent anion channel 1 (VDAC1), a major component of mitochondrial outer membrane. Knocking down VDAC1 by small interfering RNA attenuates ES-induced apoptosis, while overexpression of VDAC1 enhances the sensitivity of endothelial cells to ES. Moreover, we reveal that ES induces the reduction of hexokinase 2 (HK2), which, in turn, promotes VDAC1 phosphorylation and accumulation. Data from two-dimensional electrophoresis, immunoprecipitation, mPTP opening, and caspase-3 activation assays indicate that two serine residues of VDAC1, Ser-12 and Ser-103, can modulate VDAC1 protein level and thus the sensitivity to apoptosis stimuli. On the basis of these findings, we conclude that VDAC1 plays a vital role in modulating ES-induced endothelial cell apoptosis.


Asunto(s)
Apoptosis/fisiología , Endostatinas/fisiología , Células Endoteliales/citología , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Adenosina Trifosfato/biosíntesis , Inhibidores de la Angiogénesis/farmacología , Apoptosis/efectos de los fármacos , Secuencia de Bases , Caspasa 9/metabolismo , Línea Celular , Citocromos c/metabolismo , Endostatinas/farmacología , Células Endoteliales/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Células HeLa , Hexoquinasa/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Neovascularización Patológica , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/genética , Regulación hacia Arriba/efectos de los fármacos , Canal Aniónico 1 Dependiente del Voltaje/antagonistas & inhibidores , Canal Aniónico 1 Dependiente del Voltaje/genética
20.
Sci Rep ; 9(1): 14063, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31575916

RESUMEN

During the progression of the neurodegenerative process, mitochondria participates in several intercellular signaling pathways. Voltage-dependent anion-selective channel 1 (VDAC1) is a mitochondrial porin involved in the cellular metabolism and apoptosis intrinsic pathway in many neuropathological processes. In spinal cord injury (SCI), after the primary cell death, a secondary response that comprises the release of pro-inflammatory molecules triggers apoptosis, inflammation, and demyelination, often leading to the loss of motor functions. Here, we investigated the functional role of VDAC1 in the neurodegeneration triggered by SCI. We first determined that in vitro targeted ablation of VDAC1 by specific morpholino antisense nucleotides (MOs) clearly promotes neurite retraction, whereas a pharmacological blocker of VDAC1 oligomerization (4, 4'-diisothiocyanatostilbene-2, 2'-disulfonic acid, DIDS), does not cause this effect. We next determined that, after SCI, VDAC1 undergoes conformational changes, including oligomerization and N-terminal exposition, which are important steps in the triggering of apoptotic signaling. Considering this, we investigated the effects of DIDS in vivo application after SCI. Interestingly, blockade of VDAC1 oligomerization decreases the number of apoptotic cells without interfering in the neuroinflammatory response. DIDS attenuates the massive oligodendrocyte cell death, subserving undisputable motor function recovery. Taken together, our results suggest that the prevention of VDAC1 oligomerization might be beneficial for the clinical treatment of SCI.


Asunto(s)
Neuritas/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Médula Espinal/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/fisiología , Animales , Western Blotting , Células Cultivadas , Femenino , Técnica del Anticuerpo Fluorescente , Masculino , Microscopía Fluorescente , Ratas , Ratas Wistar , Médula Espinal/fisiopatología , Traumatismos de la Médula Espinal/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA