Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Neuropharmacology ; 196: 108360, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-33122030

RESUMEN

Mitochondria are essential for neuronal survival and function, and mitochondrial dysfunction plays a critical role in the pathological development of Parkinson's disease (PD). Mitochondrial quality control is known to contribute to the survival of dopaminergic (DA) neurons, with mitophagy being a key regulator of the quality control system. In this study, we show that mitophagy is impaired in the substantia nigra pars compacta (SNc) of the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. Treatment with the sigma-1 receptor (Sig 1R) agonist 2-morpholin-4-ylethyl 1-phenylcyclohexane-1-carboxylate (PRE-084) reduced loss of DA neurons, restored motor ability and MPTP-induced damage to mitophagy activity in the SNc of PD-like mice. Additionally, knockdown of Sig 1R in SH-SY5Y DA cells inhibited mitophagy and enhanced 1-methyl-4-phenylpyridinium ion (MPP+) neurotoxicity, whereas application of the Sig 1R selective agonist SKF10047 promoted clearance of damaged mitochondria. Moreover, knockdown of Sig 1R in SH-SY5Y cells resulted in decreased levels of p-ULK1 (Unc-51 Like Autophagy Activating Kinase 1) (Ser555), p-TBK1 (TANK Binding Kinase 1) (Ser172), p-ubiquitin (Ub) (Ser65), Parkin recruitment, and stabilization of PTEN-induced putative kinase 1 (PINK1) in mitochondria. The present data provide the first evidence for potential roles of PINK1/Parkin in Sig 1R-modulated mitophagy in DA neurons.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Mitocondrias/metabolismo , Mitofagia/genética , Trastornos Parkinsonianos/metabolismo , Proteínas Quinasas/metabolismo , Receptores sigma/genética , Ubiquitina-Proteína Ligasas/metabolismo , 1-Metil-4-fenilpiridinio/toxicidad , Animales , Homólogo de la Proteína 1 Relacionada con la Autofagia/efectos de los fármacos , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Línea Celular , Neuronas Dopaminérgicas/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Ratones , Mitocondrias/efectos de los fármacos , Mitofagia/efectos de los fármacos , Morfolinas/farmacología , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/patología , Porción Compacta de la Sustancia Negra/efectos de los fármacos , Porción Compacta de la Sustancia Negra/metabolismo , Porción Compacta de la Sustancia Negra/patología , Fenazocina/análogos & derivados , Fenazocina/farmacología , Fosforilación , Proteínas Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Estabilidad Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Receptores sigma/agonistas , Receptores sigma/metabolismo , Transducción de Señal , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Sustancia Negra/patología , Ubiquitina/efectos de los fármacos , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/efectos de los fármacos , Receptor Sigma-1
3.
Nutrients ; 12(9)2020 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-32859116

RESUMEN

One of the characteristic features of aging is the progressive loss of muscle mass, a nosological syndrome called sarcopenia. It is also a pathologic risk factor for many clinically adverse outcomes in older adults. Therefore, delaying the loss of muscle mass, through either boosting muscle protein synthesis or slowing down muscle protein degradation using nutritional supplements could be a compelling strategy to address the needs of the world's aging population. Here, we review the recently identified properties of docosahexaenoic acid (DHA). It was shown to delay muscle wasting by stimulating intermediate oxidative stress and inhibiting proteasomal degradation of muscle proteins. Both the ubiquitin-proteasome and the autophagy-lysosome systems are modulated by DHA. Collectively, growing evidence indicates that DHA is a potent pharmacological agent that could improve muscle homeostasis. Better understanding of cellular proteolytic systems associated with sarcopenia will allow us to identify novel therapeutic interventions, such as omega-3 polyunsaturated fatty acids, to treat this disease.


Asunto(s)
Autofagia/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Lisosomas/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Sarcopenia/tratamiento farmacológico , Ubiquitina/efectos de los fármacos , Envejecimiento , Humanos
4.
Neuropharmacology ; 162: 107828, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31654703

RESUMEN

Cholinergic dysfunction plays a critical role in a number of disease states, and the loss of functional muscarinic acetylcholine receptors plays a key role in disease pathogenesis. Therefore, preventing receptor downregulation would maintain functional receptor number, and be predicted to alleviate symptoms. However, the molecular mechanism(s) underlying muscarinic receptor downregulation are currently unknown. Here we demonstrate that the M2 muscarinic receptor undergoes rapid lysosomal proteolysis, and this lysosomal trafficking is facilitated by ubiquitination of the receptor. Importantly, we show that this trafficking is driven specifically by ESCRT mediated involution. Critically, we provide evidence that disruption of this process leads to a re-routing of the trafficking of the M2 receptor away from the lysosome and into recycling pathway, and eventually back to the plasma membrane. This study is the first to identify the process by which the M2 muscarinic acetylcholine receptor undergoes endocytic sorting, and critically reveals a regulatory checkpoint that represents a target to pharmacologically increase the number of functional muscarinic receptors within the central nervous system.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Lisosomas/metabolismo , Neuronas/metabolismo , Receptor Muscarínico M2/metabolismo , Ubiquitina/metabolismo , Animales , Carbacol/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Cloroquina/farmacología , Agonistas Colinérgicos/farmacología , Regulación hacia Abajo , Complejos de Clasificación Endosomal Requeridos para el Transporte/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Endosomas/ultraestructura , Ganglios Espinales/citología , Regulación de la Expresión Génica , Células HEK293 , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/ultraestructura , Microscopía Confocal , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Transporte de Proteínas/efectos de los fármacos , Proteolisis , Ratas , Receptor Muscarínico M2/efectos de los fármacos , Receptor Muscarínico M2/genética , Transfección , Ubiquitina/efectos de los fármacos , Ubiquitinación
5.
Cell Death Dis ; 10(10): 692, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31534131

RESUMEN

The cytokine TNF promotes inflammation either directly by activating the MAPK and NF-κB signaling pathways, or indirectly by triggering cell death. A20 is a potent anti-inflammatory molecule, and mutations in the gene encoding A20 are associated with a wide panel of inflammatory pathologies, both in human and in the mouse. Binding of TNF to TNFR1 triggers the NF-κB-dependent expression of A20 as part of a negative feedback mechanism preventing sustained NF-κB activation. Apart from acting as an NF-κB inhibitor, A20 is also well-known for its ability to counteract the cytotoxic potential of TNF. However, the mechanism by which A20 mediates this function and the exact cell death modality that it represses have remained incompletely understood. In the present study, we provide in vitro and in vivo evidences that deletion of A20 induces RIPK1 kinase-dependent and -independent apoptosis upon single TNF stimulation. We show that constitutively expressed A20 is recruited to TNFR1 signaling complex (Complex I) via its seventh zinc finger (ZF7) domain, in a cIAP1/2-dependent manner, within minutes after TNF sensing. We demonstrate that Complex I-recruited A20 protects cells from apoptosis by stabilizing the linear (M1) ubiquitin network associated to Complex I, a process independent of its E3 ubiquitin ligase and deubiquitylase (DUB) activities and which is counteracted by the DUB CYLD, both in vitro and in vivo. In absence of linear ubiquitylation, A20 is still recruited to Complex I via its ZF4 and ZF7 domains, but this time protects the cells from death by deploying its DUB activity. Together, our results therefore demonstrate two distinct molecular mechanisms by which constitutively expressed A20 protect cells from TNF-induced apoptosis.


Asunto(s)
Receptores Tipo I de Factores de Necrosis Tumoral/efectos adversos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/uso terapéutico , Ubiquitina/efectos de los fármacos , Animales , Apoptosis , Humanos , Ratones , Transducción de Señal , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/farmacología
6.
Microb Pathog ; 132: 362-368, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31054366

RESUMEN

Duck Tembusu virus (DTMUV) is a newly emerging pathogenic flavivirus that has caused massive economic losses to the duck industry in China. The cellular factors required for DTMUV replication have been poorly studied. The ubiquitin-proteasome system (UPS), the major intracellular proteolytic pathway, mediates diverse cellular processes, including endocytosis and signal transduction, which may be involved in the entry of virus. In the present study, we explored the interplay between DTMUV replication and the UPS in BHK-21 cells and found that treatment with proteasome inhibitor (MG132 and lactacystin) significantly decreased the DTMUV progency at the early infection stage. We further revealed that inhibition of the UPS mainly occurs on the level of viral protein expression and RNA transcription. In addition, using specific siRNAs targeting ubiquitin reduces the production of viral progeny. In the presence of MG132 the staining for the envelope protein of DTMUV was dramatically reduced in comparison with the untreated control cells. Overall, our observations reveal an important role of the UPS in multiple steps of the DTMUV infection cycle and identify the UPS as a potential drug target to modulate the impact of DTMUV infection.


Asunto(s)
Flavivirus/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Replicación Viral/fisiología , Acetilcisteína/análogos & derivados , Acetilcisteína/antagonistas & inhibidores , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Patos , Flavivirus/efectos de los fármacos , Flavivirus/patogenicidad , Técnicas de Silenciamiento del Gen , Leupeptinas/antagonistas & inhibidores , Enfermedades de las Aves de Corral/virología , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , ARN Interferente Pequeño , Transfección , Ubiquitina/efectos de los fármacos , Ubiquitina/genética , Proteínas del Envoltorio Viral , Internalización del Virus
7.
Expert Opin Ther Pat ; 28(12): 919-937, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30449221

RESUMEN

INTRODUCTION: Ubiquitin-proteasome system (UPS) has been validated as a novel anticancer drug target in the past 20 years. The UPS contains two distinct steps: ubiquitination of a substrate protein by ubiquitin activating enzyme (E1), ubiquitin conjugating enzyme (E2), and ubiquitin ligase (E3), and substrate degradation by the 26S proteasome complex. The E3 enzyme is the central player in the ubiquitination step and has a wide range of specific substrates in cancer cells, offering great opportunities for discovery and development of selective drugs. Areas covered: This review summarizes the recent advances in small molecule inhibitors of E1s, E2s, and E3s, with a focus on the latest patents (from 2015 to 2018) of E3 inhibitors and modulators. Expert opinion: One strategy to overcome limitations of current 20S proteasome inhibitors is to discover inhibitors of the upstream key components of the UPS, such as E3 enzymes. E3s play important roles in cancer development and determine the specificity of substrate ubiquitination, offering novel target opportunities. E3 modulators could be developed by rational design, natural compound or library screening, old drug repurposes, and application of other novel technologies. Further understanding of mechanisms of E3-substrate interaction will be essential for discovering and developing next-generation E3 inhibitors as effective anticancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores de Proteasoma/farmacología , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Animales , Diseño de Fármacos , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Patentes como Asunto , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/efectos de los fármacos , Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/efectos de los fármacos , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/efectos de los fármacos
8.
J Mol Biol ; 430(17): 2857-2872, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-29864443

RESUMEN

REV1 is a DNA damage tolerance protein and encodes two ubiquitin-binding motifs (UBM1 and UBM2) that are essential for REV1 functions in cell survival under DNA-damaging stress. Here we report the first solution and X-ray crystal structures of REV1 UBM2 and its complex with ubiquitin, respectively. Furthermore, we have identified the first small-molecule compound, MLAF50, that directly binds to REV1 UBM2. In the heteronuclear single quantum coherence NMR experiments, peaks of UBM2 but not of UBM1 are significantly shifted by the addition of ubiquitin, which agrees to the observation that REV1 UBM2 but not UBM1 is required for DNA damage tolerance. REV1 UBM2 interacts with hydrophobic residues of ubiquitin such as L8 and L73. NMR data suggest that MLAF50 binds to the same residues of REV1 UBM2 that interact with ubiquitin, indicating that MLAF50 can compete with the REV1 UBM2-ubiquitin interaction orthosterically. Indeed, MLAF50 inhibited the interaction of REV1 UBM2 with ubiquitin and prevented chromatin localization of REV1 induced by cisplatin in U2OS cells. Our results structurally validate REV1 UBM2 as a target of a small-molecule inhibitor and demonstrate a new avenue to targeting ubiquitination-mediated protein interactions with a chemical tool.


Asunto(s)
Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Nucleotidiltransferasas/química , Nucleotidiltransferasas/metabolismo , Éteres Fenílicos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Ubiquitina/química , Ubiquitina/metabolismo , Secuencia de Aminoácidos , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Cromatina/química , Cristalografía por Rayos X , ADN/química , ADN/metabolismo , Daño del ADN , Humanos , Modelos Moleculares , Proteínas Nucleares/efectos de los fármacos , Nucleotidiltransferasas/efectos de los fármacos , Osteosarcoma/metabolismo , Osteosarcoma/patología , Unión Proteica , Conformación Proteica , Dominios Proteicos , Células Tumorales Cultivadas , Ubiquitina/efectos de los fármacos , Ubiquitinación
9.
Viruses ; 9(11)2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29160853

RESUMEN

Epstein-Barr virus (EBV) is closely associated with several lymphomas (endemic Burkitt lymphoma, Hodgkin lymphoma and nasal NK/T-cell lymphoma) and epithelial cancers (nasopharyngeal carcinoma and gastric carcinoma). To maintain its persistence in the host cells, the virus manipulates the ubiquitin-proteasome system to regulate viral lytic reactivation, modify cell cycle checkpoints, prevent apoptosis and evade immune surveillance. In this review, we aim to provide an overview of the mechanisms by which the virus manipulates the ubiquitin-proteasome system in EBV-associated lymphoid and epithelial malignancies, to evaluate the efficacy of proteasome inhibitors on the treatment of these cancers and discuss potential novel viral-targeted treatment strategies against the EBV-associated cancers.


Asunto(s)
Infecciones por Virus de Epstein-Barr/tratamiento farmacológico , Linfoma/tratamiento farmacológico , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Inhibidores de Proteasoma/uso terapéutico , Animales , Apoptosis , Linfoma de Burkitt/tratamiento farmacológico , Linfoma de Burkitt/virología , Carcinoma/tratamiento farmacológico , Carcinoma/virología , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/virología , Antígenos Nucleares del Virus de Epstein-Barr , Herpesvirus Humano 4/patogenicidad , Enfermedad de Hodgkin/tratamiento farmacológico , Enfermedad de Hodgkin/virología , Humanos , Linfoma/virología , Ratones , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/virología , Inhibidores de Proteasoma/administración & dosificación , Ubiquitina/efectos de los fármacos
10.
BMC Cancer ; 17(1): 376, 2017 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-28549433

RESUMEN

BACKGROUND: In view of the fact that JS-K might regulate ubiquitin E3 ligase and that ubiquitin E3 ligase plays an important role in the mechanism of CRPC formation, the goal was to investigate the probable mechanism by which JS-K regulates prostate cancer cells. METHODS: Proliferation inhibition by JS-K on prostate cancer cells was examined usingCCK-8 assays. Caspase 3/7 activity assays and flow cytometry were performed to examine whether JS-K induced apoptosis in prostate cancer cells. Western blotting and co-immunoprecipitation analyses investigated JS-K's effects on the associated apoptosis mechanism. Real time-PCR and Western blotting were performed to assess JS-K's effect on transcription of specific AR target genes. Western blotting was also performed to detect Siah2 and AR protein concentrations and co-immunoprecipitation to detect interactions of Siah2 and AR, NCoR1 and AR, and p300 and AR. RESULTS: JS-K inhibited proliferation and induced apoptosis in prostate cancer cells. JS-K increased p53 and Mdm2 concentrations and regulated the caspase cascade reaction-associated protein concentrations. JS-K inhibited transcription of AR target genes and down-regulated PSA protein concentrations. JS-K inhibited Siah2 interactions and also inhibited the ubiquitination of AR. With further investigation, JS-K was found to stabilize AR and NCoR1 interactions and diminish AR and p300 interactions. CONCLUSIONS: The present results suggested that JS-K might have been able to inhibit proliferation and induce apoptosis via regulation of the ubiquitin-proteasome degradation pathway, which represented a promising platform for the development of new compounds for PCa treatments.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Compuestos Azo/farmacología , Óxido Nítrico , Piperazinas/farmacología , Neoplasias de la Próstata/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Profármacos/farmacología , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ubiquitina/efectos de los fármacos
11.
J Neurosci ; 36(6): 1930-41, 2016 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-26865616

RESUMEN

Failure to clear amyloid-ß (Aß) from the brain is in part responsible for Aß brain accumulation in Alzheimer's disease (AD). A critical protein for clearing Aß across the blood-brain barrier is the efflux transporter P-glycoprotein (P-gp) in the luminal plasma membrane of the brain capillary endothelium. P-gp is reduced at the blood-brain barrier in AD, which has been shown to be associated with Aß brain accumulation. However, the mechanism responsible for P-gp reduction in AD is not well understood. Here we focused on identifying critical mechanistic steps involved in reducing P-gp in AD. We exposed isolated rat brain capillaries to 100 nm Aß40, Aß40, aggregated Aß40, and Aß42. We observed that only Aß40 triggered reduction of P-gp protein expression and transport activity levels; this occurred in a dose- and time-dependent manner. To identify the steps involved in Aß-mediated P-gp reduction, we inhibited protein ubiquitination, protein trafficking, and the ubiquitin-proteasome system, and monitored P-gp protein expression, transport activity, and P-gp-ubiquitin levels. Thus, exposing brain capillaries to Aß40 triggers ubiquitination, internalization, and proteasomal degradation of P-gp. These findings may provide potential therapeutic targets within the blood-brain barrier to limit P-gp degradation in AD and improve Aß brain clearance. SIGNIFICANCE STATEMENT: The mechanism reducing blood-brain barrier P-glycoprotein (P-gp) in Alzheimer's disease is poorly understood. In the present study, we focused on defining this mechanism. We demonstrate that Aß40 drives P-gp ubiquitination, internalization, and proteasome-dependent degradation, reducing P-gp protein expression and transport activity in isolated brain capillaries. These findings may provide potential therapeutic avenues within the blood-brain barrier to limit P-gp degradation in Alzheimer's disease and improve Aß brain clearance.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Péptidos beta-Amiloides/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Ubiquitina/efectos de los fármacos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Animales , Transporte Biológico Activo/efectos de los fármacos , Capilares/efectos de los fármacos , Capilares/metabolismo , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Masculino , Ratas , Ratas Sprague-Dawley , Ubiquitinación/efectos de los fármacos
12.
Cancer Lett ; 356(2 Pt B): 762-72, 2015 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-25449439

RESUMEN

Sarcoma is a rare form of cancer that differs from the much more common carcinomas because it occurs in a distinct type of tissue. Many patients of sarcoma have poor response to chemotherapy and an increased risk for local recurrence. Arsenic trioxide (ATO) is used to treat certain types of leukemia. Recently, data have revealed that ATO induces sarcoma cell death in several types of solid tumor cell lines. In the present study, we investigated whether ATO induces cancer cell death and elucidated the underlying anti-cancer mechanisms. Our results showed that ATO caused concentration- and time-dependent cell death in human osteosarcoma and fibrosarcoma cells. The types of cell death that were induced by ATO were primarily autophagy and apoptosis. Furthermore, ATO activated p38, JNK and AMPK and inhibited the Akt/mTOR signaling pathways. Specifically, we found that ATO induced endoplasmic reticulum (ER) stress and suppressed proteasome activation in two types of sarcoma cell lines. However, the level of proteasome inhibition in osteosarcoma cells was lower than in fibrosarcoma cells. Thus, we used combined treatment with ATO and a proteasome inhibitor to examine the antitumor activity in fibrosarcoma cells. The data indicated showed that the combination treatment of ATO and MG132 (a proteasome inhibitor) resulted in synergistic cytotoxicity. In a fibrosarcoma xenograft mouse model, the combined treatment significantly reduced tumor progression. Immunohistochemical studies revealed that combined treatment induced autophagy and apoptosis. In summary, our results suggest a potential clinical application of ATO in sarcoma therapy and that combined treatment with a proteasome inhibitor can increase the therapeutic efficacy.


Asunto(s)
Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Autofagia/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Óxidos/farmacología , Complejo de la Endopetidasa Proteasomal/química , Sarcoma/patología , Ubiquitina/metabolismo , Animales , Antineoplásicos/farmacología , Trióxido de Arsénico , Western Blotting , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Sarcoma/tratamiento farmacológico , Sarcoma/metabolismo , Transducción de Señal/efectos de los fármacos , Células Tumorales Cultivadas , Ubiquitina/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Toxicology ; 322: 43-50, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24801902

RESUMEN

Parkin is an E3 ubiquitin ligase involved in the elimination of damaged mitochondria. Ubiquitination of mitochondrial substrates by Parkin results in proteasomal as well as lysosomal degradation of mitochondria, the latter of which is executed by the autophagy machinery and is called as mitophagy (mitochondrial autophagy). The aim of this study is to examine the possible role of Parkin against cardiotoxicity elicited by arsenic trioxide (ATO) exposure in HL-1 mouse atrial cardiomyocytes. HL-1 cells were administered 1-10µM ATO for up to 24h, and the involvements of apoptosis, and the ubiquitin-proteasome and autophagy-lysosome systems (UPS and ALS) were examined. ATO dose-dependently reduced mitochondrial membrane potentials (ΔΨm) in HL-1 cells, indicating that ATO works as a mitochondrial toxin in these cells. Apoptosis was evident in cells exposed to more than 6µM ATO for 24h. Levels of Parkin in mitochondria-rich fractions were increased, suggesting the recruitment of Parkin to mitochondria. Ubiquitination of the voltage-dependent anion channel1 (VDAC1), a substrate of Parkin, was also proved by immunoprecipitation. Accumulation of ubiquitinated proteins including both K48- and K63-lineages was observed in HL-1 cells after ATO exposure, implying an increased demand for proteasomal as well as lysosomal degradation of cellular proteins. Although UPS was activated by ATO as proved by increased proteasomal activity, only slight activation of the ALS marker LC3 was observed, suggesting differential reactions of UPS and ALS to ATO toxicity. The abrogation of UPS by the proteasome inhibitor bortezomib significantly sensitized HL-1 cells to ATO toxicity, showing the contribution of UPS to the maintenance of cellular homeostasis during ATO exposure. Taken together, our results reveal the activation of Parkin as well as UPS during ATO exposure in HL-1 cardiomyocytes, which contributes to the maintenance of mitochondrial as well as cellular homeostasis.


Asunto(s)
Cardiopatías/inducido químicamente , Cardiopatías/prevención & control , Homeostasis/efectos de los fármacos , Mitocondrias Cardíacas/efectos de los fármacos , Óxidos/toxicidad , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Ubiquitina-Proteína Ligasas/fisiología , Ubiquitina/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Arsenicales , Autofagia/efectos de los fármacos , Western Blotting , Ácidos Borónicos/farmacología , Bortezomib , Inmunoprecipitación , Lisosomas/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Microscopía Fluorescente , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Pirazinas/farmacología , Canal Aniónico 1 Dependiente del Voltaje/biosíntesis , Canal Aniónico 1 Dependiente del Voltaje/genética
14.
J Oral Pathol Med ; 43(4): 250-7, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24020947

RESUMEN

Cetuximab (Erbitux, C225) is a chimeric monoclonal antibody that binds to the extracellular domain of epidermal growth factor receptor (EGFR), inhibiting tumor growth, invasion, angiogenesis and metastasis. However, the mechanisms underlying the effect of Cetuximab in human oral squamous cell carcinoma (OSCC) remain unclear. Here, we report that Cetuximab modulates EGFR protein stability through the ubiquitin/proteasome pathway, resulting in the inhibition of human OSCC growth. Cetuximab significantly inhibited the migration and invasion of human OSCC cells by blocking epithelial/mesenchymal transition (EMT) and the AKT and ERK pathways. Furthermore, Cetuximab-inhibited cell growth by modulating the expression of integrin ß5. Taken together, these results provide novel insights into the mechanism of Cetuximab action and suggest potential therapeutic strategies for OSCC.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Carcinoma de Células Escamosas/patología , Receptores ErbB/antagonistas & inhibidores , Neoplasias de la Lengua/patología , Actinas/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/toxicidad , Carcinoma de Células Escamosas/secundario , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cetuximab , Inhibidores de Cisteína Proteinasa/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Cadenas beta de Integrinas/efectos de los fármacos , Leupeptinas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Invasividad Neoplásica , Neovascularización Patológica/patología , Proteína Oncogénica v-akt/antagonistas & inhibidores , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ubiquitina/efectos de los fármacos
15.
Cold Spring Harb Perspect Med ; 3(2): a013573, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23378599

RESUMEN

Bacterial pathogens produce protein toxins to influence host-pathogen interactions and tip the outcome of these encounters toward the benefit of the pathogen. Protein toxins modify host-specific targets through posttranslational modifications (PTMs) or noncovalent interactions that may inhibit or activate host cell physiology to benefit the pathogen. Recent advances have identified new PTMs and host targets for toxin action. Understanding the mechanisms of toxin action provides a basis to develop vaccines and therapies to combat bacterial pathogens and to develop new strategies to use toxin derivatives for the treatment of human disease.


Asunto(s)
Toxinas Bacterianas/farmacología , Actinas/efectos de los fármacos , Infecciones Bacterianas/fisiopatología , Toxinas Bacterianas/clasificación , Proteínas Activadoras de GTPasa/efectos de los fármacos , Interacciones Huésped-Patógeno/fisiología , Humanos , Biosíntesis de Proteínas/efectos de los fármacos , Procesamiento Proteico-Postraduccional/fisiología , Proteínas SNARE/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ubiquitina/efectos de los fármacos
16.
Neurotox Res ; 21(2): 185-94, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21773851

RESUMEN

Dysfunction of mitochondria, the ubiquitin proteasome system (UPS), and lysosomes are believed to contribute to the pathogenesis of Parkinson's disease (PD). If it were possible to rescue functionally compromised, but still viable neurons early in the disease process, this would slow the rate of neurodegeneration. Here, we used a catecholaminergic neuroblastoma cell line (SH-SY5Y) as a model of susceptible neurons in PD. To identify a target early in the cell death process that was common to all neurodegenerative processes linked with PD, cells were exposed to toxins that mimic cell death mechanisms associated with PD. The sub-cellular abnormalities that occur shortly after toxin exposure were determined. 3 h of exposure to either naphthazarin, to inhibit lysosomal function, Z-Ile-Glu(OBu(t))-Ala-Leu-H (PSI), to inhibit the UPS, or rotenone, to inhibit mitochondrial complex I, caused depolarisation of the mitochondrial membrane potential (2.5-fold, twofold, and 4.6-fold change, respectively compared to vehicle), suggesting impaired mitochondrial function. Following 24 h exposure to the same toxins, UPS and lysosomal function were also impaired, and ubiquitin levels were increased. Thus, following exposure to toxins that mimic three important, but disparate cell death mechanisms associated with PD, catecholaminergic cells initially experience mitochondrial dysfunction, which is then followed by abnormalities in UPS and lysosomal function. Thus, mitochondrial dysfunction is an early event in cell stress. We suggest that, in patients with PD, the surviving cells of the substantia nigra pars compacta are most susceptible to mitochondrial impairment. Thus, targeting the mitochondria may be useful for slowing the progression of neurodegeneration in PD.


Asunto(s)
Lisosomas/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Enfermedad de Parkinson Secundaria/inducido químicamente , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Ubiquitina/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Mitocondrias/metabolismo , Naftoquinonas/toxicidad , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Rotenona/toxicidad , Factores de Tiempo
17.
Diabetes ; 60(1): 227-38, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20980462

RESUMEN

OBJECTIVE: The islet in type 2 diabetes is characterized by ß-cell apoptosis, ß-cell endoplasmic reticulum stress, and islet amyloid deposits derived from islet amyloid polypeptide (IAPP). Toxic oligomers of IAPP form intracellularly in ß-cells in humans with type 2 diabetes, suggesting impaired clearance of misfolded proteins. In this study, we investigated whether human-IAPP (h-IAPP) disrupts the endoplasmic reticulum-associated degradation/ubiquitin/proteasome system. RESEARCH DESIGN AND METHODS: We used pancreatic tissue from humans with and without type 2 diabetes, isolated islets from h-IAPP transgenic rats, isolated human islets, and INS 832/13 cells transduced with adenoviruses expressing either h-IAPP or a comparable expression of rodent-IAPP. Immunofluorescence and Western blotting were used to detect polyubiquitinated proteins and ubiquitin carboxyl-terminal hydrolase L1 (UCH-L1) protein levels. Proteasome activity was measured in isolated rat and human islets. UCH-L1 was knocked down by small-interfering RNA in INS 832/13 cells and apoptosis was evaluated. RESULTS: We report accumulation of polyubiquinated proteins and UCH-L1 deficiency in ß-cells of humans with type 2 diabetes. These findings were reproduced by expression of oligomeric h-IAPP but not soluble rat-IAPP. Downregulation of UCH-L1 expression and activity to reproduce that caused by h-IAPP in ß-cells induced endoplasmic reticulum stress leading to apoptosis. CONCLUSIONS: Our results indicate that defective protein degradation in ß-cells in type 2 diabetes can, at least in part, be attributed to misfolded h-IAPP leading to UCH-L1 deficiency, which in turn further compromises ß-cell viability.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Células Secretoras de Insulina/fisiología , Polipéptido Amiloide de los Islotes Pancreáticos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina Tiolesterasa/deficiencia , Ubiquitina/metabolismo , Animales , Autopsia , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/metabolismo , Obesidad/complicaciones , Obesidad/patología , Ratas , Ratas Transgénicas , Ubiquitina/efectos de los fármacos , Ubiquitina Tiolesterasa/efectos de los fármacos
18.
Cancer Lett ; 294(1): 82-90, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20153923

RESUMEN

The ubiquitin-proteasome system (UPS) and autophagy provide major cellular pathways for protein degradation. Since the p53 pathway controls autophagy, we investigated whether p53 regulates UPS in ovarian tumour cell lines. A reporter cell line (SKOV3-EGFPu) was established to measure UPS function against a constant genetic background. Transient expression of either wild type or mutant p53 in SKOV3-EGFPu cells reduced UPS activity as compared to vector control. These results, together with those from endogenous p53 expression in seven ovarian cancer cell lines, suggest that expression of both wild-type and mutant p53 protein impairs UPS function. Thus, p53 expression may regulate protein homeostasis by down-regulating UPS function in response to cellular stress.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina/metabolismo , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Clonación Molecular , Cartilla de ADN , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Genes p53 , Homeostasis , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Plásmidos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/genética , Inhibidores de Proteasoma , Proteína p53 Supresora de Tumor/efectos de los fármacos , Ubiquitina/antagonistas & inhibidores , Ubiquitina/efectos de los fármacos , Ubiquitina/genética , Regulación hacia Arriba
19.
J Environ Radioact ; 101(3): 230-6, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19969403

RESUMEN

These experiments were designed to investigate transcriptional effects in Atlantic salmon (Salmo salar) after exposure in vivo to ionizing gamma radiation combined with subtoxic levels of aluminum (Al) and cadmium (Cd). Juvenile fish (35 g) in freshwater with or without Al and Cd (255 microg Al/L + 6 microg Cd/L) were exposed to a 75 mGy dose of gamma-irradiation, and induced responses were compared to those of controls. The transcriptional levels of eight genes encoding proteins known to respond to stress in fish were quantified in liver of fish exposed for 5 h to gamma radiation, to Al and Cd or to the combination of Al, Cd and gamma radiation. The studied genes were caspase 3B, caspase 6A, caspase 7, p53 (apoptosis), glutathione reductase (GR), phospholipid hydroperoxide glutathione peroxidase (GSH-Px), (oxidative stress), metallothionein (MT-A) (metal stress) and ubiquitin (Ubi) (protein degradation). The results showed that gamma-irradiation alone induced significant upregulation of caspase 6A, GR, GSH-Px, MT-A and Ubi compared to the control group, while 5 h exposure to Al+Cd alone did not induce any of the studied genes compared to the control. No significant upregulation of the series of investigated genes could be observed in fish exposed to gamma-irradiation in combination with Al+Cl. In conclusion, the results suggest that the presence of Al+Cd in the water counteracted the gamma-irradiation effect by modifying the transcription of genes encoding proteins involved in the defense mechanisms against free radicals in the cells.


Asunto(s)
Rayos gamma , Metales/farmacología , Salmo salar/fisiología , Aluminio/toxicidad , Animales , Cadmio/toxicidad , Caspasas/efectos de los fármacos , Caspasas/genética , Caspasas/efectos de la radiación , Radioisótopos de Cobalto/farmacología , Daño del ADN , Exposición a Riesgos Ambientales , Agua Dulce , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Metalotioneína/efectos de los fármacos , Metalotioneína/genética , Metalotioneína/efectos de la radiación , Reacción en Cadena de la Polimerasa/métodos , Proteínas/efectos de los fármacos , Proteínas/genética , Proteínas/efectos de la radiación , ARN/sangre , ARN/efectos de los fármacos , ARN/genética , Salmo salar/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/efectos de la radiación , Ubiquitina/efectos de los fármacos , Ubiquitina/genética , Ubiquitina/efectos de la radiación
20.
Crit Care ; 13(4): 311, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19691815

RESUMEN

Endotoxic shock is a serious systemic inflammatory response to an external biological stressor. The responsiveness of NF-kappaB is built upon rapid protein modification and degradation involving the ubiquitin proteasome pathway. Using transgenic mice, we have obtained in vivo evidence that interference with this pathway can alleviate the symptoms of toxic shock. We posit that administration of proteasome inhibitors may enhance the survival of patients with septic shock.


Asunto(s)
Inhibidores de Proteasas/farmacología , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Choque Séptico/tratamiento farmacológico , Ubiquitina/efectos de los fármacos , Animales , Ratones , Ratones Transgénicos , Mieloma Múltiple/fisiopatología , FN-kappa B/antagonistas & inhibidores , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/uso terapéutico , Choque Séptico/fisiopatología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...