Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Acta Biochim Biophys Sin (Shanghai) ; 56(5): 697-708, 2024 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-38591121

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most prevalent and deadly cancers in the world, which is frequently diagnosed at a late stage. HCC patients have a poor prognosis due to the lack of an efficacious therapeutic strategy. Approved drug repurposing is a way for accelerating drug discovery and can significantly reduce the cost of drug development. Carfilzomib (CFZ) is a second-generation proteasome inhibitor, which is highly efficacious against multiple myeloma and has been reported to possess potential antitumor activities against multiple cancers. However, the underlying mechanism of CFZ on HCC is still unclear. Here, we show that CFZ inhibits the proliferation of HCC cells through cell cycle arrest at the G2/M phase and suppresses the migration and invasion of HCC cells by inhibiting epithelial-mesenchymal transition. We also find that CFZ promotes reactive oxygen species production to induce endoplasmic reticulum (ER) stress and activate JNK/p38 MAPK signaling in HCC cells, thus inducing cell death in HCC cells. Moreover, CFZ significantly inhibits HCC cell growth in a xenograft mouse model. Collectively, our study elucidates that CFZ impairs mitochondrial function and activates ER stress and JNK/p38 MAPK signaling, thus inhibiting HCC cell and tumor growth. This indicates that CFZ has the potential as a therapeutic drug for HCC.


Asunto(s)
Apoptosis , Carcinoma Hepatocelular , Estrés del Retículo Endoplásmico , Neoplasias Hepáticas , Oligopéptidos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Oligopéptidos/farmacología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Ratones , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proliferación Celular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Desnudos , Movimiento Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Ratones Endogámicos BALB C
2.
Zhongguo Yi Liao Qi Xie Za Zhi ; 45(5): 542-545, 2021 Sep 30.
Artículo en Zh | MEDLINE | ID: mdl-34628769

RESUMEN

In recent years, with the wide application of magnetic resonance imaging (MRI) equipment in clinical practice, the quality of the equipment causes adverse events, which put pressure on manufacturers, at the same time, it puts forward higher requirements for medical device supervisors. In order to help the medical device supervisors to clarify the key points of verification, this paper analyzes the main risk points in the production process of the product according to the medical device good manufacturing practice(GMP), and puts forward the suggestions for field verification, which has practical significance for the submission of verification efficiency.


Asunto(s)
Comercio , Imagen por Resonancia Magnética
3.
Arch Toxicol ; 93(11): 3367-3383, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31563988

RESUMEN

The primary liver cancer (PLC) is one of the leading causes of cancer-related death worldwide. The predominant form of PLC is hepatocellular carcinoma (HCC), which accounts for about 85% of all PLC. Artemisinin (ART) was clinically used as anti-malarial agents. Recently, it was demonstrated to inhibit cell growth and migration in multiple cancer types. However, the molecular mechanism underlying these anti-cancer activity remains largely unknown. Herein, it is discovered that ART dramatically suppresses HCC cell growth in vitro through arresting cell cycle progression, and represses cell migration and invasion via regulating N-cadherin-Snail-E-cadherin axis. In addition, the disruption of cellular bioenergetics contributed to ART-caused cell growth, migration and invasion inhibition. Moreover, ART (100 mg/kg, intraperitoneally) substantially inhibits HCC xenograft growth in vivo. Importantly, Hippo-YAP signal transduction is remarkably inactivated in HCC cells upon ART administration. Collectively, these data reveal a novel mechanism of ART in regulating HCC cell growth, migration, and invasion, which indicates that ART could be considered as a potential drug for the treatment of HCC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antineoplásicos Fitogénicos/farmacología , Artemisininas/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Vía de Señalización Hippo , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones Desnudos , Invasividad Neoplásica , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Señalizadoras YAP
4.
Cell Mol Life Sci ; 72(24): 4807-24, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26363553

RESUMEN

Lon protease is a nuclear DNA-encoded mitochondrial enzyme highly conserved throughout evolution, involved in the degradation of damaged and oxidized proteins of the mitochondrial matrix, in the correct folding of proteins imported in mitochondria, and in the maintenance of mitochondrial DNA. Lon expression is induced by various stimuli, including hypoxia and reactive oxygen species, and provides protection against cell stress. Lon down-regulation is associated with ageing and with cell senescence, while up-regulation is observed in tumour cells, and is correlated with a more aggressive phenotype of cancer. Lon up-regulation contributes to metabolic reprogramming observed in cancer, favours the switch from a respiratory to a glycolytic metabolism, helping cancer cell survival in the tumour microenvironment, and contributes to epithelial to mesenchymal transition. Silencing of Lon, or pharmacological inhibition of its activity, causes cell death in various cancer cells. Thus, Lon can be included in the growing class of proteins that are not responsible for oncogenic transformation, but that are essential for survival and proliferation of cancer cells, and that can be considered as a new target for development of anticancer drugs.


Asunto(s)
Envejecimiento/genética , Proteínas Mitocondriales/fisiología , Neoplasias/genética , Estrés Oxidativo , Proteasa La/fisiología , Envejecimiento/patología , Proliferación Celular , Supervivencia Celular , Secuencia Conservada , Evolución Molecular , Regulación de la Expresión Génica , Humanos , Redes y Vías Metabólicas , Mitocondrias/enzimología , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Modelos Biológicos , Neoplasias/patología , Proteasa La/genética , Proteasa La/metabolismo
5.
J Biol Chem ; 289(37): 25737-49, 2014 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-25074939

RESUMEN

MERTK, a member of the TAM (TYRO3, AXL, and MERTK) receptor tyrosine kinases, has complex and diverse roles in cell biology. On the one hand, knock-out of MERTK results in age-dependent autoimmunity characterized by failure of apoptotic cell clearance, while on the other, MERTK overexpression in cancer drives classical oncogene pathways leading to cell transformation. To better understand the interplay between cell transformation and efferocytosis, we stably expressed MERTK in human MCF10A cells, a non-tumorigenic breast epithelial cell line devoid of endogenous MERTK. While stable expression of MERTK in MCF10A resulted in enhanced motility and AKT-mediated chemoprotection, MERTK-10A cells did not form stable colonies in soft agar, or enhance proliferation compared with parental MCF10A cells. Concomitant to chemoresistance, MERTK also stimulated efferocytosis in a gain-of-function capacity. However, unlike AXL, MERTK activation was highly dependent on apoptotic cells, suggesting MERTK may preferentially interface with phosphatidylserine. Consistent with this idea, knockdown of MERTK in breast cancer cells MDA-MB 231 reduced efferocytosis, while transient or stable expression of MERTK stimulated apoptotic cell clearance in all cell lines tested. Moreover, human breast cancer cells with higher endogenous MERTK showed higher levels of efferocytosis that could be blocked by soluble TAM receptors. Finally, through MERTK, apoptotic cells induced PD-L1 expression, an immune checkpoint blockade, suggesting that cancer cells may adopt MERTK-driven efferocytosis as an immune suppression mechanism for their advantage. These data collectively identify MERTK as a significant link between cancer progression and efferocytosis, and a potentially unrealized tumor-promoting event when MERTK is overexpressed in epithelial cells.


Asunto(s)
Neoplasias de la Mama/genética , Transformación Celular Neoplásica/genética , Células Epiteliales/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Apoptosis/genética , Neoplasias de la Mama/patología , Movimiento Celular/genética , Células Epiteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Fagocitosis/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Tirosina Quinasa c-Mer , Tirosina Quinasa del Receptor Axl
6.
Research (Wash D C) ; 6: 0175, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37333972

RESUMEN

Interorganelle contacts and communications are increasingly recognized to play a vital role in cellular function and homeostasis. In particular, the mitochondria-endoplasmic reticulum (ER) membrane contact site (MAM) is known to regulate ion and lipid transfer, as well as signaling and organelle dynamics. However, the regulatory mechanisms of MAM formation and their function are still elusive. Here, we identify mitochondrial Lon protease (LonP1), a highly conserved mitochondrial matrix protease, as a new MAM tethering protein. The removal of LonP1 substantially reduces MAM formation and causes mitochondrial fragmentation. Furthermore, deletion of LonP1 in the cardiomyocytes of mouse heart impairs MAM integrity and mitochondrial fusion and activates the unfolded protein response within the ER (UPRER). Consequently, cardiac-specific LonP1 deficiency causes aberrant metabolic reprogramming and pathological heart remodeling. These findings demonstrate that LonP1 is a novel MAM-localized protein orchestrating MAM integrity, mitochondrial dynamics, and UPRER, offering exciting new insights into the potential therapeutic strategy for heart failure.

7.
Redox Biol ; 54: 102366, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35728303

RESUMEN

Despite the evidences of elevated expression of Mer tyrosine kinase (MerTK) in multiple human cancers, mechanisms underlying the oncogenic roles of MerTK in hepatocellular carcinoma (HCC) remains undefined. We explored the functional effects of MerTK and N-Glycosylated MerTK on HCC cell survival and tumor growth. Here, we show that MerTK ablation increases reactive oxygen species (ROS) production and promotes the switching from glycolytic metabolism to oxidative phosphorylation in HCC cells, thus suppressing HCC cell proliferation and tumor growth. MerTK is N-glycosylated in HCC cells at asparagine 294 and 454 that stabilizes MerTK to promote oncogenic transformation. Moreover, we observed that nuclear located non-glycosylated MerTK is indispensable for survival of HCC cells under stress. Pathologically, tissue microarray (TMA) data indicate that MerTK is a pivotal prognostic factor for HCC. Our data strongly support the roles of MerTK N-glycosylation in HCC tumorigenesis and suggesting N-glycosylation inhibition as a potential HCC therapeutic strategy.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinogénesis/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Glicosilación , Humanos , Neoplasias Hepáticas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Tirosina Quinasa c-Mer/genética , Tirosina Quinasa c-Mer/metabolismo
8.
Cell Death Dis ; 12(3): 251, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33674562

RESUMEN

Ciclopirox (CPX) is an antifungal drug that has recently been reported to act as a potential anticancer drug. However, the effects and underlying molecular mechanisms of CPX on glioblastoma multiforme (GBM) remain unknown. Bortezomib (BTZ) is the first proteasome inhibitor-based anticancer drug approved to treat multiple myeloma and mantle cell lymphoma, as BTZ exhibits toxic effects on diverse tumor cells. Herein, we show that CPX displays strong anti-tumorigenic activity on GBM. Mechanistically, CPX inhibits GBM cellular migration and invasion by reducing N-Cadherin, MMP9 and Snail expression. Further analysis revealed that CPX suppresses the expression of several key subunits of mitochondrial enzyme complex, thus leading to the disruption of mitochondrial oxidative phosphorylation (OXPHOS) in GBM cells. In combination with BTZ, CPX promotes apoptosis in GBM cells through the induction of reactive oxygen species (ROS)-mediated c-Jun N-terminal kinase (JNK)/p38 mitogen-activated protein kinase (MAPK) signaling. Moreover, CPX and BTZ synergistically activates nuclear factor kappa B (NF-κB) signaling and induces cellular senescence. Our findings suggest that a combination of CPX and BTZ may serve as a novel therapeutic strategy to enhance the anticancer activity of CPX against GBM.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bortezomib/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Ciclopirox/farmacología , Glioblastoma/tratamiento farmacológico , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Glioblastoma/enzimología , Glioblastoma/patología , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Fosforilación Oxidativa/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
EBioMedicine ; 41: 408-419, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30826359

RESUMEN

BACKGROUND: Endoplasmic reticulum (ER) oxidoreductin-1α (Ero1α) and protein disulfide isomerase (PDI) constitute the pivotal pathway of oxidative protein folding, and are highly expressed in many cancers. However, whether targeting the functional interplay between Ero1α and PDI could be a new approach for cancer therapy remains unknown. METHODS: We performed wound healing assays, transwell migration and invasion assays and xenograft assays to assess cell migration, invasion and tumorigenesis; gel filtration chromatography, oxygen consumption assay and in cells folding assays were used to detect Ero1α-PDI interaction and Ero1α oxidase activity. FINDINGS: Here, we report that elevated expression of Ero1α is correlated with poor prognosis in human cervical cancer. Knockout of ERO1A decreases the growth, migration and tumorigenesis of cervical cancer cells, through downregulation of the H2O2-correlated epithelial-mesenchymal transition. We identify that the conserved valine (Val) 101 of Ero1α is critical for Ero1α-PDI complex formation and Ero1α oxidase activity. Val101 of Ero1α is specifically involved in the recognition of PDI catalytic domain. Mutation of Val101 results in a reduced ER, retarded oxidative protein folding and decreased H2O2 levels in the ER of cervical cancer cells and further impairs cell migration, invasion, and tumor growth. INTERPRETATION: Our study identifies the critical residue of Ero1α for recognizing PDI, which underlines the molecular mechanism of oxidative protein folding for tumorigenesis and provides a proof-of-concept for cancer therapy by targeting Ero1α-PDI interaction. FUND: This work was supported by National Key R&D Program of China, National Natural Science Foundation of China, and Youth Innovation Promotion Association, CAS.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Oxidorreductasas/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Neoplasias del Cuello Uterino/patología , Animales , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Femenino , Humanos , Peróxido de Hidrógeno/metabolismo , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Mutagénesis Sitio-Dirigida , Oxidorreductasas/antagonistas & inhibidores , Oxidorreductasas/genética , Pronóstico , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Tasa de Supervivencia , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/mortalidad
10.
Cancer Lett ; 432: 132-143, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-29906487

RESUMEN

Deferoxamine (DFO) was found to modulate multiple cellular pathways involved in the growth of breast cancer, hepatocellular carcinoma, lung cancer and bladder cancer. However, the effect of DFO on esophageal squamous cell carcinoma (ESCC) remains unclear. Here, we report that DFO-treated ESCC cells show strong anti-tumorigenic properties, such as inhibition of cell proliferation, induction of cell cycle arrest, and promotion of apoptosis. Mechanistically, DFO significantly activated ERK1/2 signaling, which is reactive oxygen species (ROS)-dependent. ERK1/2 activation suppressed mitochondrial respiration and aerobic glycolysis in ESCC cells, resulting in reduced production of ATP and key precursor metabolites. Cell proliferation was functionally rescued by the ROS scavenger N-acetyl-l-cysteine (NAC) and the ERK1/2 inhibitor SCH7 72984. Additionally, our data showed that activated ERK1/2 was partially translocated to the mitochondria, which indicated that DFO-activated ERK1/2 may suppress tumor formation through inhibition of mitochondrial respiration. Moreover, the decreased c-Myc expression caused by DFO resulted in the inhibition of cell migration. Taken together, our study demonstrate that DFO activates ERK1/2 and downregulates c-Myc to perturb mitochondrial homeostasis and promote apoptosis, resulting in the novel anti-neoplastic activity of DFO in ESCC.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Deferoxamina/farmacología , Neoplasias Esofágicas/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Mitocondrias/patología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Animales , Apoptosis , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Movimiento Celular , Proliferación Celular , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Femenino , Humanos , Ratones , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Sideróforos/farmacología , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Res ; 78(11): 2813-2824, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29572227

RESUMEN

Mitochondria regulate cellular bioenergetics and redox states and influence multiple signaling pathways required for tumorigenesis. In this study, we determined that the mitochondrial translation elongation factor 4 (EF4) is a critical component of tumor progression. EF4 was ubiquitous in human tissues with localization to the mitochondria (mtEF4) and performed quality control on respiratory chain biogenesis. Knockout of mtEF4 induced respiratory chain complex defects and apoptosis, while its overexpression stimulated cancer development. In multiple cancers, expression of mtEF4 was increased in patient tumor tissues. These findings reveal that mtEF4 expression may promote tumorigenesis via an imbalance in the regulation of mitochondrial activities and subsequent variation of cellular redox. Thus, dysregulated mitochondrial translation may play a vital role in the etiology and development of diverse human cancers.Significance: Dysregulated mitochondrial translation drives tumor development and progression. Cancer Res; 78(11); 2813-24. ©2018 AACR.


Asunto(s)
Metabolismo Energético/fisiología , Mitocondrias/metabolismo , Neoplasias/metabolismo , Factores de Elongación de Péptidos/metabolismo , Células A549 , Apoptosis/fisiología , Carcinogénesis/metabolismo , Línea Celular Tumoral , Células HeLa , Células Hep G2 , Humanos , Células K562 , Oxidación-Reducción , Células PC-3 , Transducción de Señal/fisiología
12.
Oncol Rep ; 37(4): 2237-2244, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28350087

RESUMEN

Paclitaxel induces apoptosis in a variety of cancer cells. However, the mechanism of paclitaxel inducing apoptosis in human esophageal squamous cell carcinoma (ESCC) remains to be defined. In this study, we found that paclitaxel-induced apoptosis by increasing the relevant apoptosis protein expression and the release of cytochrome c via downregulation of signal transducer and activator of transcription 3 (STAT3) and phospho-STAT3 (Ser727). In addition, paclitaxel treatment of ESCC cells EC-1 and Eca-109 led to marked mitochondrial membrane potential depolarization and significantly increasing of reactive oxygen species. Moreover, paclitaxel treatment resulted in the inhibition of mitochondrial respiration. In conclusion, our findings reveal that paclitaxel induced apoptosis in both EC-1 and Eca-109 cells through the reduction of STAT3 and phospho­STAT3 (Ser727) level, and suggest that paclitaxel may be of therapeutic potential in the treatment of ESCC through the induction of mitochondrial apoptosis in ESCC cells.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/metabolismo , Mitocondrias/efectos de los fármacos , Paclitaxel/farmacología , Factor de Transcripción STAT3/metabolismo , Serina/metabolismo , Apoptosis , Carcinoma de Células Escamosas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Neoplasias Esofágicas/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Fosforilación/efectos de los fármacos
13.
Biosci Rep ; 37(3)2017 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-28465355

RESUMEN

The natural small molecule compound: 2,3,5,6-tetramethylpyrazine (TMP), is a major component of the Chinese medicine Chuanxiong, which has wide clinical applications in dilating blood vessels, inhibiting platelet aggregation and treating thrombosis. Recent work suggests that TMP is also an antitumour agent. Despite its chemotherapeutic potential, the mechanism(s) underlying TMP action are unknown. Herein, we demonstrate that TMP binds to mitochondrial transcription factor A (TFAM) and blocks its degradation by the mitochondrial Lon protease. TFAM is a key regulator of mtDNA replication, transcription and transmission. Our previous work showed that when TFAM is not bound to DNA, it is rapidly degraded by the ATP-dependent Lon protease, which is essential for mitochondrial proteostasis. In cultured cells, TMP specifically blocks Lon-mediated degradation of TFAM, leading to TFAM accumulation and subsequent up-regulation of mtDNA content in cells with substantially low levels of mtDNA. In vitro protease assays show that TMP does not directly inhibit mitochondrial Lon, rather interacts with TFAM and blocks degradation. Pull-down assays show that biotinylated TMP interacts with TFAM. These findings suggest a novel mechanism whereby TMP stabilizes TFAM and confers resistance to Lon-mediated degradation, thereby promoting mtDNA up-regulation in cells with low mtDNA content.


Asunto(s)
ADN Mitocondrial/efectos de los fármacos , Proteínas de Unión al ADN/genética , Dosificación de Gen/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Proteínas Mitocondriales/genética , Pirazinas/farmacología , Factores de Transcripción/genética , Regulación hacia Arriba/efectos de los fármacos , Línea Celular Tumoral , Replicación del ADN/efectos de los fármacos , Células HCT116 , Células HeLa , Humanos , Péptido Hidrolasas/genética , Transcripción Genética/efectos de los fármacos
14.
Biosci Rep ; 37(6)2017 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-29026004

RESUMEN

The aim of the present study was to explore the effects of oxidative stress induced by CoCl2 and H2O2 on the regulation of bioenergetics of esophageal squamous cell carcinoma (ESCC) cell line TE-1 and analyze its underlying mechanism. Western blot results showed that CoCl2 and H2O2 treatment of TE-1 cells led to significant reduction in mitochondrial respiratory chain complex subunits expression and increasing intracellular reactive oxygen species (ROS) production. We further found that TE-1 cells treated with CoCl2, a hypoxia-mimicking reagent, dramatically reduced the oxygen consumption rate (OCR) and increased the extracellular acidification rate (ECAR). However, H2O2 treatment decreased both the mitochondrial respiration and aerobic glycolysis significantly. Moreover, we found that H2O2 induces apoptosis in TE-1 cells through the activation of PARP, Caspase 3, and Caspase 9. Therefore, our findings indicate that CoCl2 and H2O2 could cause mitochondrial dysfunction by up-regulation of ROS and regulating the cellular bioenergy metabolism, thus affecting the survival of tumor cells.


Asunto(s)
Carcinoma de Células Escamosas/patología , Metabolismo Energético/fisiología , Neoplasias Esofágicas/patología , Estrés Oxidativo/fisiología , Apoptosis/fisiología , Carcinoma de Células Escamosas/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Línea Celular Tumoral , Supervivencia Celular/fisiología , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas de Esófago , Humanos , Mitocondrias/patología , Consumo de Oxígeno/fisiología , Especies Reactivas de Oxígeno/metabolismo
15.
Yi Chuan ; 28(2): 148-52, 2006 Feb.
Artículo en Zh | MEDLINE | ID: mdl-16520308

RESUMEN

We report for the first time in China, the characterization of a special abnormal inv(Y) with dual-color fluorescence in situ hybridization (D-FISH). We also study the mechanism of the formation of chromosome Y inversion and its relationship with the phenotype of recurrent spontaneous abortion. Using biotin-11-dUTP-labeled Yp11.3 breakpoint probe (No. 889) and CY3-labeled Yq12 heterochromatic region DNA probe (PY3.4), we performed D-FISH on a balanced translocation carrier with a [46, XY(90%)/46, X, inv(Y)(p11.3; q12)] karyotype determined by G-banding karyotyping, whose wife had recurrent miscarriages. The result of D-FISH shows that the percentage of cells with abnormal karyotypes is about 22%, higher than that determined by G-banding analysis. Moreover, besides the type of inversion diagnosed by G-banding, there are the two other abnormal karyotypes, including a type of chromatid inversion, which is difficult to be detected by conventional G-banding technique. The present of the three types of inversion confirms that the breakpoints of inv(Y) are heterogeneous. D-FISH is a powerful tool for the detection of chromosomal inversion due to its sensitivity and specificity.


Asunto(s)
Inversión Cromosómica/genética , Cromosomas Humanos Y/genética , Adulto , Bandeo Cromosómico , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Masculino
16.
Oncotarget ; 7(10): 11609-24, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26820294

RESUMEN

Mitochondrial transcription factor A (TFAM) is essential for the replication, transcription and maintenance of mitochondrial DNA (mtDNA). The role of TFAM in non-small cell lung cancer (NSCLC) remains largely unknown. Herein, we report that downregulation of TFAM in NSCLC cells resulted in cell cycle arrest at G1 phase and significantly blocked NSCLC cell growth and migration through the activation of reactive oxygen species (ROS)-induced c-Jun amino-terminal kinase(JNK)/p38 MAPK signaling and decreased cellular bioenergetics. We further found that TFAM downregulation in NSCLC cells led to increased apoptotic cell death and enhanced the sensitivity of NSCLC cells to cisplatin. Tissue microarray (TMA) data showed that elevated expression of TFAM was related to the histological grade and TNM stage of NSCLC patients. We also demonstrated that TFAM is an independent prognostic factor for overall survival of NSCLC patients. Taken together, our findings suggest that TFAM could serve as a potential diagnostic biomarker and molecular target for the treatment of NSCLC, as well as for prediction of the effectiveness of chemotherapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Pulmonares/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Mitocondriales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/metabolismo , Animales , Carcinogénesis , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Femenino , Células HEK293 , Xenoinjertos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Transducción de Señal , Factores de Transcripción/genética , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Yi Chuan ; 27(2): 185-9, 2005 Mar.
Artículo en Zh | MEDLINE | ID: mdl-15843342

RESUMEN

To study the technique of fluorescence in situ hybridization (FISH) and its application value in the diagnosis of sex chromosomal count abnormality in ovarian carcinoma cell. Biotin labeled alpha satellite X chromosome DNA(pBamX7) probe was hybridized with pre-treated slides of ovarian carcinoma cell interphase nucleus in 18 cases of ovarian carcinoma specimens. The slides were treated with Avidin-FITC and Anti-avidin, amplified with an additional layer and counter-stained with PI in antifade solution. The hybridization signals as well as interphase nucleus settings were observed with WIB filters under fluorescence microscope Olympus AX-70, and the number of interphase nucleus in the ovarian carcinoma cell was counted. It was observed under the microscope that the Biotin labeled pBamX7 probe showed green hybridization signals. Cytoplasm counter-stained with PI showed reddish orange. Increased chrosome X copy number was observed in 11/18(61%) ovarian carcinoma specimens, of which the rest 7 (39%) had no increase of chrosome X copy number. Gain of X chrosome had a certain incidence in ovarian cancers, which played a role in the recurrence and development of ovarian cancers. Its significance needs further investigation.


Asunto(s)
Cromosomas Humanos X/genética , Hibridación Fluorescente in Situ/métodos , Aberraciones Cromosómicas Sexuales , Adulto , Cistadenocarcinoma Mucinoso/genética , Cistadenocarcinoma Mucinoso/patología , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología , Femenino , Humanos , Interfase/genética , Microscopía Fluorescente , Persona de Mediana Edad , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Células Tumorales Cultivadas
18.
Oncotarget ; 6(35): 37792-807, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26473374

RESUMEN

Crk is the prototypical member of a class of Src homology 2 (SH2) and Src homology 3 (SH3) domain-containing adaptor proteins that positively regulate cell motility via the activation of Rac1 and, in certain tumor types such as GBM, can promote cell invasion and metastasis by mechanisms that are not well understood. Here we demonstrate that Crk, via its phosphorylation at Tyr251, promotes invasive behavior of tumor cells, is a prominent feature in GBM, and correlating with aggressive glioma grade IV staging and overall poor survival outcomes. At the molecular level, Tyr251 phosphorylation of Crk is negatively regulated by Abi1, which competes for Crk binding to Abl and attenuates Abl transactivation. Together, these results show that Crk and Abi1 have reciprocal biological effects and act as a molecular rheostat to control Abl activation and cell invasion. Finally, these data suggest that Crk Tyr251 phosphorylation regulate invasive cell phenotypes and may serve as a biomarker for aggressive GBM.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Biomarcadores de Tumor/metabolismo , Movimiento Celular , Proteínas del Citoesqueleto/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patología , Proteínas Proto-Oncogénicas c-abl/metabolismo , Proteínas Proto-Oncogénicas c-crk/metabolismo , Apoptosis , Sitios de Unión , Western Blotting , Proliferación Celular , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioblastoma/mortalidad , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica , Fenotipo , Fosforilación , Pronóstico , Transducción de Señal , Tasa de Supervivencia , Análisis de Matrices Tisulares , Células Tumorales Cultivadas , Cicatrización de Heridas
19.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 20(5): 433-5, 2003 Oct.
Artículo en Zh | MEDLINE | ID: mdl-14556200

RESUMEN

OBJECTIVE: To investigate the pathogenesis of male sterility in 48,XXYY syndrome patient. METHODS: The peripheral lymphocyte was detected by dual-color fluorescence in situ hybridization. The bioptic testicular tissues were pathologically sectioned and ultra-thin sections were examined by electron-microscopy. RESULTS: The pathological findings revealed extremely severe dysgenesis of the badly damaged testicular tissue. Only a few convoluted seminiferous tubules were found, in which no spermatogenic cell or sperm of any range could be viewed. The ultrastructural observations showed the thickened interstitial vascular walls of the testicular tissue and severe hyperplasia of the collagen fibers in the basilemma and lumens of the blood vessels. CONCLUSION: The structure of the testicle in the 48,XXYY syndrome patient has severe fibrous hyperplasia, leading to the non-specific thickening of the barrier and serious damage to the blood-testis barrier, which in turn produce significant disturbance and pathological changes in the process of the spermatogenic cell formation. The whole interrelated loops account mainly for the male sterility.


Asunto(s)
Cromosomas Humanos X/genética , Cromosomas Humanos Y/genética , Linfocitos/metabolismo , Aberraciones Cromosómicas Sexuales , Testículo/ultraestructura , Adulto , Humanos , Hibridación Fluorescente in Situ/métodos , Infertilidad Masculina/genética , Infertilidad Masculina/patología , Masculino , Microscopía Electrónica , Testículo/metabolismo , Testículo/patología
20.
Yi Chuan ; 25(3): 271-5, 2003 May.
Artículo en Zh | MEDLINE | ID: mdl-15639868

RESUMEN

The objective of the work is to study the technique of dual-color fluorescence in situ hybridization(D-FISH) and its application value in the diagnosis of sex chromosomal count abnormality Klinefelter syndrome and establish an experimental approach to metaphase chromosome and interphase nucleus FISH technique. Biotin labeled alpha satellite X-chromosome DNA(pBamX7) probe and Digoxigenin labeled Y-chromosome long arm terminal repetitive sequence (pY3.4) probe were hybridized with pre-treated slides of peripheral blood chromosome and interphase nucleus in 19 cases of Klinefelter syndrome specimens. After being washed,the slides were treated with Avidin-FITC,Rhodamine-FITC and Anti-avidin,amplified with an additional layer and counter-stained with DAPI in an antifade solution. The hybridization signals,chromosomal or interphase nucleus settings were observed respectively with WIB, WIG and WU filters under fluorescence microscope Olympus AX-70,and the number of metaphase chromosome and interphase nucleus in the peripheral blood was counted. It was observed under the microscope that the Biotin labeled pBamX7 probe showed 2 green hybridization signals and that the Digoxigenin labeled pY3.4 probe showed 1 red hybridization signal. Chromosome or interphase nucleus counter-stained with DAPI showed blue. The average signal rate of chromosome and interphase nucleus hybridization was 95.89% and 95% respectively,significantly higher than the normal control (2.75%). Karyotype 47,XXY was confirmed,which agrees with the chromosomal findings. One case showed mosaic nuclei. XXY chromosome hybridization signal rate was 92% and XY hybridization signal rate was 6.7%, higher than the normal control rate of 4.17%. FISH is a valuable technique in diagnosing sex chromosomal count abnormality Klinefelter syndrome with the merits of fast speed, high sensitivity, strong signal,low background and multiple color. Therefore, FISH technique can find wide application and potential in prenatal diagnosis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA