Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int J Cancer ; 143(9): 2213-2224, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29761481

RESUMEN

TGF-ß plays a central role in mediating epithelial-mesenchymal transition (EMT) by activating the Smad pathway. In addition, accumulating evidence suggests that TGF-ß-induced EMT is NF-κB-dependent in various cancer types. However, it is largely unclear if NF-κB mediates TGF-ß-induced EMT in breast cancer, and if this mediation occurs, the regulatory mechanisms are unknown. In our study, we found that TGF-ß activates the NF-κB pathway. Inhibition of NF-κB signaling markedly abrogates TGF-ß-induced EMT. By studying the regulatory mechanism of TGF-ß-induced NF-κB signaling, we found that lncRNA NKILA was upregulated by TGF-ß and was essential for the negative feedback regulation of the NF-κB pathway. Accordingly, overexpression of NKILA significantly reduced TGF-ß-induced tumor metastasis in vivo. Consistent with the results from mice, the expression of NKILA was negatively correlated with EMT phenotypes in clinical breast cancer samples. Collectively, our study indicated that the NKILA-mediated negative feedback affects TGF-ß-induced NF-κB activation and that NKILA may be a therapeutic molecule in breast cancer metastasis via inhibition of EMT.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Cadherinas/metabolismo , Transición Epitelial-Mesenquimal , FN-kappa B/metabolismo , ARN Largo no Codificante/genética , Factor de Crecimiento Transformador beta/farmacología , Animales , Apoptosis , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Cadherinas/genética , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , FN-kappa B/genética , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Hepatology ; 61(5): 1730-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25605636

RESUMEN

UNLABELLED: Valproic acid (VPA) is widely used to treat epilepsy, migraine, chronic headache, bipolar disorder, and as adjuvant chemotherapy, but potentially causes idiosyncratic liver injury. Alpers-Huttenlocher syndrome (AHS), a neurometabolic disorder caused by mutations in mitochondrial DNA polymerase gamma (POLG), is associated with an increased risk of developing fatal VPA hepatotoxicity. However, the mechanistic link of this clinical mystery remains unknown. Here, fibroblasts from 2 AHS patients were reprogrammed to induced pluripotent stem cells (iPSCs) and then differentiated to hepatocyte-like cells (AHS iPSCs-Hep). Both AHS iPSCs-Hep are more sensitive to VPA-induced mitochondrial-dependent apoptosis than controls, showing more activated caspase-9 and cytochrome c release. Strikingly, levels of both soluble and oligomeric optic atrophy 1, which together keep cristae junctions tight, are reduced in AHS iPSCs-Hep. Furthermore, POLG mutation cells show reduced POLG expression, mitochondrial DNA (mtDNA) amount, mitochondrial adenosine triphosphate production, as well as abnormal mitochondrial ultrastructure after differentiation to hepatocyte-like cells. Superoxide flashes, spontaneous bursts of superoxide generation, caused by opening of the mitochondrial permeability transition pore (mPTP), occur more frequently in AHS iPSCs-Hep. Moreover, the mPTP inhibitor, cyclosporine A, rescues VPA-induced apoptotic sensitivity in AHS iPSCs-Hep. This result suggests that targeting mPTP opening could be an effective method to prevent hepatotoxicity by VPA in AHS patients. In addition, carnitine or N-acetylcysteine, which has been used in the treatment of VPA-induced hepatotoxicity, is able to rescue VPA-induced apoptotic sensitivity in AHS iPSCs-Hep. CONCLUSION: AHS iPSCs-Hep are more sensitive to the VPA-induced mitochondrial-dependent apoptotic pathway, and this effect is mediated by mPTP opening. Toxicity models in genetic diseases using iPSCs enable the evaluation of drugs for therapeutic targets.


Asunto(s)
Anticonvulsivantes/efectos adversos , Apoptosis , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Esclerosis Cerebral Difusa de Schilder/complicaciones , Células Madre Pluripotentes Inducidas , Proteínas de Transporte de Membrana Mitocondrial/fisiología , Ácido Valproico/efectos adversos , Células Cultivadas , Humanos , Poro de Transición de la Permeabilidad Mitocondrial
3.
Arch Insect Biochem Physiol ; 83(4): 195-210, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23740663

RESUMEN

Sf-caspase-1 is the principal effector caspase in Spodoptera frugiperda cells. Like the caspases in other organisms, Sf-caspase-1 is processed by upstream caspases to form an active heterotetramer composed of the p19 and p12 subunits. The regulation of active caspases is crucial for cellular viability. In mammal cells, the subunits and the active form of caspase-3 were rapidly degraded relative to its proenzyme form. In the present study, the S. frugiperda Sf9 cells were transiently transfected with plasmids encoding different fragments of Sf-caspase-1: the pro-Sf-caspase-1 (p37), a prodomain deleted fragment (p31), a fragment containing the large subunit and the prodomain (p25), the large subunit (p19), and the small subunit (p12). Flow cytometry and Western blot analysis revealed that p12, p19, and p25 were unstable in the transfected cells, in contrast to p37 and p31. Lactacystin, a proteasome inhibitor, increased the accumulation of the p19 and p12 subunits, suggesting that the degradation is performed by the ubiquitin-proteasome system. During the activation, the Sf-caspase-1 produces an intermediate form and then undergoes proteolytic processing to form active Sf-caspase-1. We found that both the active and the intermediate form were unstable, indicating that once activated or during its activation, the Sf-caspase-1 was unstable.


Asunto(s)
Caspasas/metabolismo , Activación Enzimática/fisiología , Spodoptera/enzimología , Acetilcisteína/análogos & derivados , Animales , Western Blotting , Clonación Molecular , Cartilla de ADN/genética , Estabilidad de Enzimas/fisiología , Citometría de Flujo , Etiquetado Corte-Fin in Situ , Indoles , Mutagénesis Sitio-Dirigida , Subunidades de Proteína/metabolismo , Células Sf9
4.
Sci Adv ; 8(15): eabk2376, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35417232

RESUMEN

Mitochondrial quality control plays an important role in maintaining mitochondrial homeostasis and function. Disruption of mitochondrial quality control degrades brain function. We found that flunarizine (FNZ), a drug whose chronic use causes parkinsonism, led to a parkinsonism-like motor dysfunction in mice. FNZ induced mitochondrial dysfunction and decreased mitochondrial mass specifically in the brain. FNZ decreased mitochondrial content in both neurons and astrocytes, without affecting the number of nigral dopaminergic neurons. In human neural progenitor cells, FNZ also induced mitochondrial depletion. Mechanistically, independent of ATG5- or RAB9-mediated mitophagy, mitochondria were engulfed by lysosomes, followed by a vesicle-associated membrane protein 2- and syntaxin-4-dependent extracellular secretion. A genome-wide CRISPR knockout screen identified genes required for FNZ-induced mitochondrial elimination. These results reveal not only a previously unidentified lysosome-associated exocytosis process of mitochondrial quality control that may participate in the FNZ-induced parkinsonism but also a drug-based method for generating mitochondria-depleted mammal cells.

5.
STAR Protoc ; 2(2): 100568, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-34151295

RESUMEN

Somatic cells can be reprogrammed into induced pluripotent stem cells (iPSCs) by defined factors. Here, we describe a protocol for imaging mitochondrial permeability transition pore (mPTP) opening in reprogramming of somatic cells using a confocal microscope. We also describe a method to sort high and low mPTP opening somatic cells by calcein fluorescence and reprogram these sorted cells to iPSCs. These protocols are also suitable for imaging mPTP opening and uncovering the mechanisms of mPTP function in other cell fate conversions. For complete details on the use and execution of this protocol, please refer to Ying et al. (2018).


Asunto(s)
Reprogramación Celular , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Animales , Células Cultivadas , Medios de Cultivo , Ratones , Ratones Transgénicos
6.
Cell Metab ; 28(6): 935-945.e5, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30174306

RESUMEN

Reprogramming of somatic cells to induced pluripotent stem cells reconfigures chromatin modifications. Whether and how this process is regulated by signals originating in the mitochondria remain unknown. Here we show that the mitochondrial permeability transition pore (mPTP), a key regulator of mitochondrial homeostasis, undergoes short-term opening during the early phase of reprogramming and that this transient activation enhances reprogramming. In mouse embryonic fibroblasts, greater mPTP opening correlates with higher reprogramming efficiency. The reprogramming-promoting function of mPTP opening is mediated by plant homeodomain finger protein 8 (PHF8) demethylation of H3K9me2 and H3K27me3, leading to reduction in their occupancies at the promoter regions of pluripotency genes. mPTP opening increases PHF8 protein levels downstream of mitochondrial reactive oxygen species production and miR-101c and simultaneously elevates levels of PHF8's cofactor, α-ketoglutarate. Our findings represent a novel mitochondria-to-nucleus pathway in cell fate determination by mPTP-mediated epigenetic regulation.


Asunto(s)
Reprogramación Celular , Fibroblastos/metabolismo , Histona Demetilasas/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/fisiología , Membranas Mitocondriales/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Factores de Transcripción/metabolismo , Animales , Células HEK293 , Humanos , Ácidos Cetoglutáricos/metabolismo , Metilación , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Especies Reactivas de Oxígeno/metabolismo
7.
Autophagy ; 13(9): 1543-1555, 2017 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-28722510

RESUMEN

Induced pluripotent stem cells (iPSCs) have fewer and immature mitochondria than somatic cells and mainly rely on glycolysis for energy source. During somatic cell reprogramming, somatic mitochondria and other organelles get remodeled. However, events of organelle remodeling and interaction during somatic cell reprogramming have not been extensively explored. We show that both SKP/SKO (Sox2, Klf4, Pou5f1/Oct4) and SKPM/SKOM (SKP/SKO plus Myc/c-Myc) reprogramming lead to decreased mitochondrial mass but with different kinetics and by divergent pathways. Rapid, MYC/c-MYC-induced cell proliferation may function as the main driver of mitochondrial decrease in SKPM/SKOM reprogramming. In SKP/SKO reprogramming, however, mitochondrial mass initially increases and subsequently decreases via mitophagy. This mitophagy is dependent on the mitochondrial outer membrane receptor BNIP3L/NIX but not on mitochondrial membrane potential (ΔΨm) dissipation, and this SKP/SKO-induced mitophagy functions in an important role during the reprogramming process. Furthermore, endosome-related RAB5 is involved in mitophagosome formation in SKP/SKO reprogramming. These results reveal a novel role of mitophagy in reprogramming that entails the interaction between mitochondria, macroautophagy/autophagy and endosomes.


Asunto(s)
Reprogramación Celular , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Mitofagia , Animales , Embrión de Mamíferos/citología , Endosomas/metabolismo , Endosomas/ultraestructura , Fibroblastos/metabolismo , Factor 4 Similar a Kruppel , Potencial de la Membrana Mitocondrial , Ratones , Mitocondrias/ultraestructura , Modelos Biológicos , Factores de Transcripción/metabolismo , Proteínas de Unión al GTP rab5/metabolismo
8.
Sci Rep ; 6: 28072, 2016 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-27321273

RESUMEN

As baculoviruses usually have a narrow insecticidal spectrum, knowing the mechanisms by which they control the host-range is prerequisite for improvement of their applications as pesticides. In this study, from supernatant of culture cells transfected with DNAs of an Autographa californica multiple nucleopolyhedrovirus (AcMNPV) mutant lacking the antiapoptotic gene p35 (vAc(∆P35)) and a cosmid representing a fragment of Spodoptera exigua nucleopolyhedrovirus (SeMNPV), a viral strain was plaque-purified and named vAcRev. vAcRev had a broader host range than either vAc(∆P35) or SeMNPV parental virus, being able to infect not only the permissive hosts of its parental viruses but also a nonpermissive host (Spodoptera litura). Genome sequencing indicated that vAcRev comprises a mixture of two viruses with different circular dsDNA genomes. One virus contains a genome similar to vAc(∆P35), while in the other viral genome, a 24.4 kbp-fragment containing 10 essential genesis replaced with a 4 kbp-fragment containing three SeMNPV genes including a truncated Se-iap3 gene. RNA interference and ectopic expression assays found that Se-iap3 is responsible for the host range expansion of vAcRev, suggesting that Se-iap3 inhibits the progression of apoptosis initiated by viral infection and promotes viral propagation in hosts both permissive and non-permissive for AcMNPV and SeMNPV.


Asunto(s)
Especificidad del Huésped/fisiología , Nucleopoliedrovirus/genética , Spodoptera/virología , Animales , Cósmidos/genética , Cósmidos/metabolismo , ADN Viral/química , ADN Viral/metabolismo , Genoma Viral/genética , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Lepidópteros/virología , Nucleopoliedrovirus/crecimiento & desarrollo , Nucleopoliedrovirus/fisiología , Interferencia de ARN , Análisis de Secuencia de ADN , Células Sf9/citología , Células Sf9/virología , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
9.
CNS Neurosci Ther ; 22(8): 648-60, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27080255

RESUMEN

BACKGROUND: Neuritic degeneration is an important early pathological step in neurodegeneration. AIM: The purpose of this study was to explore the mechanisms connecting neuritic degeneration to the functional and morphological remodeling of endoplasmic reticulum (ER) and mitochondria. METHODS: Here, we set up neuritic degeneration models by neurite cutting-induced neural degeneration in human-induced pluripotent stem cell-derived neurons. RESULTS: We found that neuritic ER becomes fragmented and forms complexes with mitochondria, which induces IP3R-dependent mitochondrial Ca(2+) elevation and dysfunction during neuritic degeneration. Furthermore, mitochondrial membrane potential is required for ER fragmentation and mitochondrial Ca(2+) elevation during neuritic degeneration. Mechanically, tightening of the ER-mitochondria associations by expression of a short "synthetic linker" and ER Ca(2+) releasing together could promote mitochondrial Ca(2+) elevation, dysfunction, and reactive oxygen species generation. CONCLUSION: Our study reveals a dynamic remodeling of the ER-mitochondria interface underlying neuritic degeneration.


Asunto(s)
Retículo Endoplásmico/fisiología , Potencial de la Membrana Mitocondrial/fisiología , Degeneración Nerviosa/fisiopatología , Neuritas/ultraestructura , Neuronas/ultraestructura , Apoptosis , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Células Cultivadas , Feto , Células HEK293 , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/fisiología , Neuritas/fisiología , Neuronas/efectos de los fármacos , Oligodesoxirribonucleótidos/farmacología , Células Madre Pluripotentes/efectos de los fármacos , Ionóforos de Protónes/farmacología , Especies Reactivas de Oxígeno
10.
Cell Metab ; 23(1): 220-6, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26549484

RESUMEN

The mechanisms of somatic cell reprogramming have been revealed at multiple levels. However, the lack of tools to monitor different reactive oxygen species (ROS) has left their distinct signals and roles in reprogramming unknown. We hypothesized that mitochondrial flashes (mitoflashes), recently identified spontaneous bursts of mitochondrial superoxide signaling, play a role in reprogramming. Here we show that the frequency of mitoflashes transiently increases, accompanied by flash amplitude reduction, during the early stages of reprogramming. This transient activation of mitoflashes at the early stage enhances reprogramming, whereas sustained activation impairs reprogramming. The reprogramming-promoting function of mitoflashes occurs via the upregulation of Nanog expression that is associated with decreases in the methylation status of the Nanog promoter through Tet2 occupancy. Together our findings provide a previously unknown role for superoxide signaling mediated epigenetic regulation in cell fate determination.


Asunto(s)
Reprogramación Celular , Proteínas de Homeodominio/metabolismo , Mitocondrias/fisiología , Animales , Células Cultivadas , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Epigénesis Genética , Fibroblastos/fisiología , Proteínas de Homeodominio/genética , Humanos , Ratones , Proteína Homeótica Nanog , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Superóxidos/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA