Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Stress Chaperones ; 27(1): 83-95, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35050463

RESUMEN

Metastasis is responsible for most of the hepatocellular carcinoma (HCC)-associated death. However, its underlying mechanism has yet to be fully elucidated. Glycolysis-derived lactate has been shown to be a powerful regulator of cancer metastasis. Heat shock protein A12A (HSPA12A) encodes a novel member of HSP70 family. We have recently demonstrated that heat shock protein A12A (HSPA12A) inhibited renal cancer cell migration by suppressing lactate output and glycolytic activity, which were mediated by unstabilizing CD147 and promoting its degradation. By striking contrast, here we demonstrated that HSPA12A promoted migration of human HCC cells. Extracellular acidification, lactate export, and glycolytic activity in HCC cells were also promoted following HSPA12A overexpression. Further analysis revealed that HSPA12A interacted with MCT4 and increased its membrane localization, thereby promoting export of lactate generated from glycolysis; this led, ultimately, to HCC cell migration. Our results revealed the opposite effect of HSPA12A on migration of renal cancer cells and that of HCC cells. Of note, in contrast to the inhibitory effect on CD147 expression in renal cancer cells, we found that HSPA12A increased CD147 expression in HCC cells, indicating that the expression of CD147 might exist heterogeneity in different cancer cell types. Taken together, we identified HSPA12A as an activator of HCC migration, a role opposite to that of renal cancer cells. Inhibiting HSPA12A might be a potential therapeutic intervention for HCC metastasis.


Asunto(s)
Carcinoma Hepatocelular , Proteínas HSP70 de Choque Térmico/metabolismo , Neoplasias Renales , Neoplasias Hepáticas , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Femenino , Proteínas de Choque Térmico , Humanos , Lactatos , Neoplasias Hepáticas/metabolismo , Masculino
2.
Toxicol Appl Pharmacol ; 434: 115798, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34793778

RESUMEN

Local anesthetics (LAs) are widely used for intraoperative anesthesia and postoperative analgesia. However, LAs (e.g. Bupivacaine) can evoke myotoxicity that closely associated to mitochondrial damage. PGC1a is a mast co-factor for mitochondrial quality control. We have recently demonstrated that PGC1a can be activated by HSPA12A in hepatocytes, suggesting a possibility that HSPA12A protects from LAs myotoxicity through activating PGC1α-mediated mitochondrial integrity. Here, we reported that HSPA12A was downregulated during Bupivacaine-induced myotoxicity in skeletal muscles of mice in vivo and C2c12 myoblast cultures in vitro. Intriguingly, overexpression of HSPA12A attenuated the Bupivacaine-induced C2c12 cell death. We also noticed that the Bupivacaine-induced decrease of glucose consumption and ATP production was improved by HSPA12A overexpression. Moreover, overexpression of HSPA12A in C2c12 cells attenuated the Bupivacaine-induced decrease of mitochondrial contents and increase of mitochondrial fragmentation. The Bupivacaine-induced reduction of PGC1α expression and nuclear localization was markedly attenuated by HSPA12A overexpression. Importantly, pretreatment with a selective PGC1α inhibitor (SR-18292) abolished the protection of HSPA12A from Bupivacaine-induced death and mitochondrial loss in C2c12 cells. Altogether, the findings indicate that downregulation of HSPA12A underlies myotoxicity of Local anesthetic agent Bupivacaine through inhibiting PGC1α-mediated Mitochondrial Integrity. Thus, HSPA12A might represent a viable strategy for preventing myotoxicity of LAs.


Asunto(s)
Bupivacaína/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Enfermedades Musculares/inducido químicamente , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Anestésicos Locales/toxicidad , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Proteínas HSP70 de Choque Térmico , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética
3.
Int Immunopharmacol ; 99: 107987, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34343936

RESUMEN

Acute lung injury (ALI) is a critical manifestation of sepsis/septic shock. Disruption of endothelial barrier function is critical for ALI pathogenesis; however, the regulation of endothelial barrier integrity remains largely unclear. Heat shock protein A12A (HSPA12A) is an atypical member of HSP70 family. We have recently demonstrated that hepatocyte HSPA12A attenuated the bacteria endotoxin (lipopolysaccharide, LPS)-induced liver injury. However, the role of HSPA12A in endothelial barrier function and ALI is unknown. Here in this study, HSPA12A showed upregulation in lungs of mice during bacteria endotoxin (lipopolysaccharide, LPS)-induced lung injury in vivo and in primary human umbilical vein endothelial cells (HUVECs) during LPS-induced barrier disruption in vitro. Knockout of HSPA12A in mice exacerbated LPS-induced ALI. Intriguingly, overexpression of HSPA12A in HUVECs attenuated the LPS-induced endothelial hyperpermeability. In line with this, HSPA12A overexpression increased VE-cadherin and decreased VEGF expression following LPS treatment in HUVECs. Also, knockout of HSPA12A enhanced the LPS-evoked pulmonary endothelial cell apoptosis in mice whereas overexpression of HSPA12A inhibited the LPS-induced death of HUVECs. The levels of ERKs and Akt phosphorylation in HUVECs were promoted by HSPA12A overexpression when cells exposed to LPS. Importantly, inhibition of either ERKs or Akt diminished the HSPA12A-induced protection from LPS-induced endothelial hyperpermeability and death. Taken together, these findings indicated that HSPA12A is a novel regulator of endothelial barrier function through both ERKs and Akt-mediated signaling. HSPA12A might represent a viable strategy for the pulmonary protection against endotoxemia challenge.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Endotelio Vascular/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Animales , Apoptosis , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Endotoxemia/inducido químicamente , Proteínas HSP70 de Choque Térmico/deficiencia , Proteínas HSP70 de Choque Térmico/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lipopolisacáridos/farmacología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
4.
BMC Anesthesiol ; 21(1): 49, 2021 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-33581727

RESUMEN

BACKGROUND: Postoperative nausea and vomiting (PONV) is one of the most frequent complications following strabismus surgery. Penehyclidine, an anticholinergic agent, is widely used as premedication. This study investigated the effect of preoperative penehyclidine on PONV in patients undergoing strabismus surgery. METHODS: In this prospective, randomized, double-blind study, patients scheduled for strabismus surgery under general anesthesia were randomly assigned to either penehyclidine (n = 114) or normal saline (n = 104) group. Penehyclidine was administrated immediately after anesthesia induction, and normal saline was substituted as control. PONV was investigated from 0 to 48 h after surgery. Intraoperative oculocardiac reflex (OCR) was also recorded. RESULTS: Compared with normal saline, penehyclidine significantly reduced PONV incidence (30.7% vs. 54.8%, P < 0.01) and mitigated PONV severity as indicated by severity scoring (P < 0.01). Compared with normal saline, penehyclidine also significantly reduced OCR incidence (57.9% vs. 77.9%, P < 0.01) and mitigated OCR severity, as indicated by the requirement for atropine rescue (77.3% vs. 90.1%, P < 0.05) and the maximum decrease of heart rate during OCR (23.1 ± 9.4 bpm vs. 27.3 ± 12.4 bpm, P < 0.05). The recovery course did not differ between groups. CONCLUSIONS: Penehyclidine administrated after anesthesia induction significantly reduced the incidence of PONV and alleviated intraoperative OCR in patients undergoing strabismus surgery. TRIAL REGISTRATION: ClinicalTrials.gov ( NCT04054479 ). Retrospectively registered August 13, 2019.


Asunto(s)
Complicaciones Intraoperatorias/prevención & control , Náusea y Vómito Posoperatorios/prevención & control , Quinuclidinas/farmacología , Reflejo Oculocardíaco/efectos de los fármacos , Estrabismo/cirugía , Adolescente , Adulto , Anciano , Niño , Preescolar , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
5.
Theranostics ; 10(19): 8573-8590, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32754264

RESUMEN

Background: Metastasis accounts for 90% of cancer-associated mortality in patients with renal cell carcinoma (RCC). However, the clinical management of RCC metastasis is challenging. Lactate export is known to play an important role in cancer cell migration. This study investigated the role of heat shock protein A12A (HSPA12A) in RCC migration. Methods: HSPA12A expression was examined in 82 pairs of matched RCC tumors and corresponding normal kidney tissues from patients by immunoblotting and immunofluorescence analyses. The proliferation of RCC cells was analyzed using MTT and EdU incorporation assays. The migration of RCC cells was evaluated by wound healing and Transwell migration assays. Extracellular acidification was examined using Seahorse technology. Protein stability was determined following treatment with protein synthesis inhibitor cycloheximide and proteasome inhibitor MG132. Mass spectrometry, immunoprecipitation, and immunoblotting were employed to examine protein-protein interactions. Results: RCC tumors from patients showed downregulation of HSPA12A, which was associated with advanced tumor node metastasis stage. Intriguingly, overexpression of HSPA12A in RCC cells inhibited migration, whereas HSPA12A knockdown had the opposite effect. Lactate export, glycolysis rate, and CD147 protein abundance were also inhibited by HSPA12A overexpression but promoted by HSPA12A knockdown. An interaction of HSPA12A with HRD1 ubiquitin E3 ligase was detected in RCC cells. Further studies demonstrated that CD147 ubiquitination and proteasomal degradation were promoted by HSPA12A overexpression whereas inhibited by HSPA12A knockdown. Notably, the HSPA12A overexpression-induced inhibition of lactate export and migration were abolished by CD147 overexpression. Conclusion: Human RCC shows downregulation of HSPA12A. Overexpression of HSPA12A in RCC cells unstabilizes CD147 through increasing its ubiquitin-proteasome degradation, thereby inhibits lactate export and glycolysis, and ultimately suppresses RCC cell migration. Our results demonstrate that overexpression of HSPA12A might represent a viable strategy for managing RCC metastasis.


Asunto(s)
Basigina/metabolismo , Carcinoma de Células Renales/patología , Regulación hacia Abajo , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Neoplasias Renales/patología , Ácido Láctico/metabolismo , Transporte Biológico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Glucólisis , Células Hep G2 , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Pronóstico , Estabilidad Proteica , Análisis de Supervivencia
6.
FEBS J ; 287(24): 5464-5477, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32128976

RESUMEN

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death. Proliferating cell nuclear antigen (PCNA) plays a pivotal role in cancer development and progression. However, the long-term dismal prognosis of HCC mandates more investigation to identify novel regulators in HCC pathogenesis. Heat-shock protein A12A (HSPA12A) encodes a novel member of the HSP70 family. Here, we report that HCC cells showed increased HSPA12A expression, and overexpression of HSPA12A promoted HCC growth and angiogenesis in mice. Gain- and loss-of-functional studies demonstrated that the proliferation of HCC HepG2 cells, as well as ß-catenin expression and nuclear translocation, was promoted by HSPA12A overexpression, but in turn suppressed by HSPA12A knockdown. HSPA12A did not impact PCNA expression; however, mass spectrometry and co-immunoprecipitation immunoblotting analysis revealed that HSPA12A directly binds to PCNA and promotes its trimerization, which is an essential functional conformation of PCNA for carcinogenesis. Importantly, PCNA inhibition by PCNA-I1 reversed the HSPA12A-mediated HepG2 cell differentiation. These findings indicate that HSPA12A is a novel regulator of HCC cell proliferation and tumor growth through binding to PCNA for its trimerization. HSPA12A inhibition might represent a viable strategy for the management of HCC in humans.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/patología , Proliferación Celular , Proteínas HSP70 de Choque Térmico/metabolismo , Neoplasias Hepáticas/patología , Neovascularización Patológica/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas HSP70 de Choque Térmico/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Ratones Desnudos , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Fosforilación , Antígeno Nuclear de Célula en Proliferación/genética , Unión Proteica , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1451-1459, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30797958

RESUMEN

Browning of white adipose tissues (WAT) is critical for a variety of physiological and pathophysiological events. Given the limited understanding in molecular control of WAT browning, further research is needed. Heat shock protein A12A (HSPA12A) is a new member of multigene Hsp70 family. This study investigated the effect of HSPA12A on the browning of WAT. WAT Browning in mice was induced by cold exposure for 5 days. We observed that nuclear HSPA12A content was increased in WAT after cold exposure, while deficiency of HSPA12A (Hspa12a-/-) promoted the cold-induced browning of WAT in mice compared to wild type (WT) littermates. Accordingly, Hspa12a-/- mice showed attenuation of body temperature drop and increase of thermogenic gene expression compared to WT mice after cold exposure. However, in vitro experiments demonstrated that HSPA12A deficiency in primary white adipocytes did not affect their browning and thermogenic gene expression. Further loss- and gain-of-HSPA12A functional studies revealed that HSPA12A deficiency promoted whereas HSPA12A overexpression impeded M2 macrophage polarization. Importantly, the conditioned medium (CM) from Hspa12a-/- bone marrow-derived macrophages (BMDMs) enhanced the browning of primary white adipocytes when compared to the CM from WT BMDMs. The data identified macrophage HSPA12A as a novel regulator of WAT browning through a paracrine mechanism. Targeting HSPA12A might provide meaningful advances for the management of browning-associated physiological events such as hypothermia adaptation and pathophysiological disorders such as obesity and cancer-related cachexia.


Asunto(s)
Adaptación Fisiológica/genética , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Metabolismo Energético/genética , Proteínas HSP70 de Choque Térmico/genética , Macrófagos/metabolismo , Adipocitos Blancos/citología , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Animales , Temperatura Corporal , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Frío , Medios de Cultivo Condicionados/farmacología , Expresión Génica , Proteínas HSP70 de Choque Térmico/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Comunicación Paracrina/genética , Cultivo Primario de Células , Células RAW 264.7
8.
Cardiovasc Res ; 115(1): 154-167, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29982352

RESUMEN

Aims: Inadequate healing after myocardial infarction (MI) leads to heart failure and fatal ventricular rupture, while optimal healing requires timely induction and resolution of inflammation. This study tested the hypothesis that heat shock protein B1 (HSPB1), which limits myocardial inflammation during endotoxemia, modulates wound healing after MI. Methods and results: To test this hypothesis, cardiomyocyte-specific HSPB1 knockout (Hspb1-/-) mice were generated using the Cre-LoxP recombination system. MI was induced by ligation of the left anterior descending coronary artery in Hspb1-/- and wild-type (WT) littermates. HSPB1 was up-regulated in cardiomyocytes of WT animals in response to MI, and deficiency of cardiomyocyte HSPB1 increased MI-induced cardiac rupture and mortality within 21 days after MI. Serial echocardiography showed more aggravated remodelling and cardiac dysfunction in Hspb1-/- mice than in WT mice at 1, 3, and 7 days after MI. Decreased collagen deposition and angiogenesis, as well as increased MMP2 and MMP9 activity, were also observed in Hspb1-/- mice compared with WT controls after MI, using immunofluorescence, polarized light microscopy, and zymographic analyses. Notably, Hspb1-/- hearts exhibited enhanced and prolonged leucocyte infiltration, enhanced expression of inflammatory cytokines, and enhanced TLR4/MyD88/NFκB activation compared with WT controls after MI. In-depth molecular analyses in both mice and primary cardiomyocytes demonstrated that cardiomyocyte-specific knockout of HSPB1 increased nuclear factor-κB (NFκB) activation, which promoted the expression of proinflammatory mediators. This led to increased leucocyte recruitment, thereby to excessive inflammation, ultimately resulting in adverse remodelling, cardiac dysfunction, and cardiac rupture following MI. Conclusion: These data suggest that HSPB1 acts as a negative regulator of NFκB-mediated leucocyte recruitment and the subsequent inflammation in cardiomyocytes. Cardiomyocyte HSPB1 is required for wound healing after MI and could be a target for myocardial repair in MI patients.


Asunto(s)
Quimiotaxis de Leucocito , Rotura Cardíaca Posinfarto/metabolismo , Proteínas de Choque Térmico/deficiencia , Leucocitos/metabolismo , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , FN-kappa B/metabolismo , Remodelación Ventricular , Animales , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteínas de Choque Térmico HSP27/deficiencia , Proteínas de Choque Térmico HSP27/genética , Rotura Cardíaca Posinfarto/inmunología , Rotura Cardíaca Posinfarto/patología , Rotura Cardíaca Posinfarto/fisiopatología , Proteínas de Choque Térmico/genética , Leucocitos/inmunología , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/genética , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/inmunología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , Neovascularización Fisiológica , Ratas Sprague-Dawley , Transducción de Señal , Cicatrización de Heridas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...