Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Ann Clin Lab Sci ; 49(1): 105-111, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30814085

RESUMEN

During a spinal cord injury (SCI), mechanical trauma rapidly leads to a blood-spinal cord barrier (BSB) disruption, neural cell damage, axonal damage, and demyelination, followed by a cascade of secondary inflammatory reactions. These inflammatory responses spread the damage to the neural cells and impair the recovery of neurological functions. In the present study, we evaluated the efficacy of simvastatin and a simvastatin-ezetimibe combination therapy in managing the endothelial inflammatory response in an SCI rat model. Adult male Sprague-Dawley rats were group-housed and SCI was induced by using the modified weight-drop method. The animals were divided into 4 groups: (1) sham group, laminectomy only (n=6); (2) no-treatment group, SCI without therapy (n=8); (3) simvastatin group (n=8), and (4) ezetimibe and simvastatin combination therapy group (n=8). A high dose (15 mg/kg) of simvastatin was given to the simvastatin group, and 10 mg/kg simvastatin and 10 mg/kg ezetimibe were given to the combination group. Neurological function was assessed using the Basso, Beattie, and Bresnahan locomotor scale score. Intercellular adhesion molecule-1 (ICAM-1) level was used as an SCI biomarker. ICAM-1 level was the highest at 72 hours after SCI in the no-treament group. The treatment groups showed significant reduction in ICAM-1 levels at 72 hours. The treatment groups, especially the combination treatment group, showed better neurological function scores. Simvastatin and simvastatin- ezetimibe all could improve the neurological function and attenuate the endothelial inflammatory response after spinal cord injury in rat.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Ezetimiba/farmacología , Inflamación/tratamiento farmacológico , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Simvastatina/farmacología , Traumatismos de la Médula Espinal/complicaciones , Animales , Anticolesterolemiantes/farmacología , Quimioterapia Combinada , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Inflamación/etiología , Inflamación/patología , Masculino , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función
2.
Taiwan J Obstet Gynecol ; 58(1): 122-127, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30638465

RESUMEN

OBJECTIVE: Sleep deprivation (SD) adversely affects female reproductive function. In this study, we investigated the role of glucocorticoids in ovarian development in sleep deprived rats. MATERIALS AND METHODS: Female rats were subjected to SD for 1-4 days. Concentrations of serum estradiol and corticosterone were assessed. Betamethasone (BET) and/or recombinant human follicle-stimulating hormone (FSH) were administered to 21-day-old female rats for 2 days to evaluate ovarian status for follicular development. Intact preantral follicles were mechanically dissected from the rat's ovaries and cultured for 72 h with or without FSH in the presence or absence of BET to evaluate follicular development. RESULTS: SD led to a significant difference in serum estradiol concentrations between the sham and SD groups, and corticosterone concentrations were significantly elevated in groups with more than 2 days of SD (P < 0.05). FSH stimulated ovarian growth in immature rats, whereas BET inhibited ovarian development caused by the FSH treatment. Treatment of the preantral follicles with FSH induced an increase in both follicle size and follicular cell number, while follicular cell differentiation was accompanied by enhanced inhibin-α and connexin 43 expression. Inhibition of FSH-stimulated follicular growth through BET treatment exhibited a dose-dependent reciprocal trend; as the BET dose increased (0.001-1 µg/mL), preantral follicular growth decreased. This decrease was associated with a decrease in follicular cell numbers and suppression of a proliferating cell nuclear antigen, inhibin-α, and connexin 43 expression. CONCLUSION: The findings suggest that the adverse effects of SD may inhibit follicular development during ovarian hyperstimulation by corticosterone elevation in rat.


Asunto(s)
Corticosterona/sangre , Estradiol/sangre , Folículo Ovárico/efectos de los fármacos , Privación de Sueño/sangre , Animales , Betametasona/administración & dosificación , Betametasona/farmacología , Femenino , Hormona Folículo Estimulante/farmacología , Glucocorticoides/sangre , Glucocorticoides/farmacología , Folículo Ovárico/crecimiento & desarrollo , Folículo Ovárico/metabolismo , Distribución Aleatoria , Ratas
3.
Medicine (Baltimore) ; 96(46): e8696, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29145306

RESUMEN

Some patients with hepatitis C virus (HCV) infections who fail to achieve sustained virological responses (SVRs) after interferon (IFN) therapy do not develop hepatocellular carcinoma (HCC). Risk stratification of these patients may help identify those who would benefit most from treatment with direct-acting antivirals (DAAs).A total of 552 HCV-infected patients with non-SVR status were enrolled. Laboratory data before and after IFN treatment were analyzed to determine the relationship of changes in serum markers with development of HCC during the 7-year study period.HCC developed in 93 patients. The risk factors for HCC were pre-existing liver cirrhosis, low hemoglobin level at baseline, low pretreatment platelet count, high post-treatment alpha-fetoprotein (AFP) level (≥15 ng/mL), and high post-treatment Fibrosis 4 (FIB4) index (>3.25). For patients without pre-existing cirrhosis, those with high post-treatment AFP level and FIB4 index had the highest risk of HCC (1 year: 6.7%; 3 years: 10.9%; 5 years: 29.7%), followed by those with high post-treatment AFP level and low post-treatment FIB4 index (5 years: 25%), and those with low post-treatment AFP level and high post-treatment FIB4 index (1 year: 3.7%; 3 years: 5.2%; 5 years: 10.6%). The risk was even lower for patients with low post-treatment AFP level and FIB4 index (1 year: 0%; 3 years: 0.4%; 5 years: 2.5%). None of the patients with FIB4 indexes consistently below 1.45 developed HCC.The combined use of post-treatment AFP level and FIB4 index was useful for risk stratification of HCV-infected patients with non-SVR status after IFN therapy. These data may help clinicians to identify patients who most urgently need DAA treatment.


Asunto(s)
Antivirales/uso terapéutico , Carcinoma Hepatocelular/etiología , Hepatitis C Crónica/sangre , Hepatitis C Crónica/complicaciones , Interferones/uso terapéutico , Neoplasias Hepáticas/etiología , Respuesta Virológica Sostenida , Biomarcadores/sangre , Estudios de Cohortes , Femenino , Hepatitis C Crónica/tratamiento farmacológico , Hepatitis C Crónica/virología , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Estudios Retrospectivos
4.
Biomed Rep ; 7(5): 416-422, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29181154

RESUMEN

Pluchea indica (L.) Less. is a perennial plant known for its versatile uses in traditional medicine. Previous findings have shown that the extracts of Pluchea indica possess significant anti-inflammatory, anti-ulcer and anti-tuberculosis activity. The aim of this study was to demonstrate the anticancer activity of the hexane fraction of P. indica root extract (H-PIRE) in human glioblastoma cells using flow cytometric and western blot analysis. The results shoewd that, H-PIRE suppressed the growth of glioblastoma cells in a dose-dependent manner. H-PIRE treatment markedly decreased the population of cells in S and G2/M phases. The significant upregulation of acidic vesicular organelle (AVO) was detected during H-PIRE treatment. The expression levels of microtubule-associated light chain 3-II (LC3-II) protein, phosphorylated JNK and phosphorylated p38 were significantly increased, confirming the occurrence of autophagy during the process. Finally, the combination treatment of H-PIRE and LY294002, a pan PI3K inhibitor, further decreased cell viability, suggesting an additive anticancer effect. Taken together, our results suggest that H-PIRE suppresses the proliferation of glioblastoma cells by inducing cell cycle arrest and autophagy.

5.
PLoS One ; 12(8): e0182204, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28797039

RESUMEN

BACKGROUND AND AIMS: A recent meta-analysis revealed that the genotype PNPLA3 rs738409 GG is associated with a higher risk of hepatic steatosis (HS) in Caucasian patients with chronic hepatitis C (CHC). However, controversial results were found regarding Asian populations. Furthermore, previous studies have shown a negative association between interferon lambda 3 (IFNL3) rs12979860 CC and HS in Caucasian CHC patients, but there have been no reports indicating any such association in Asian populations. In this study, then, we investigated the association of PNPLA3 and IFNL3 polymorphisms with HS in Asian CHC patients. METHODS: We enrolled consecutive CHC patients who underwent liver biopsy prior to antiviral therapy. We excluded those patients with decompensated liver disease, any co-existing chronic liver disease, or HIV or HBV co-infection. RESULTS: 1080 CHC patients were enrolled, and HS was found in 453 (41.9%) patients. The frequency distribution of the G allele was significantly associated with HS (P<0.001), and this conferred a higher risk to G allele homozygotes (OR: 2.06, 95% CI: 1.46-2.88, P <0.001) than to G allele carriers (OR: 1.98, 95% CI: 1.52-2.58, P<0.001). There was a borderline significant difference in the prevalence of HS in rs12979860 CC versus non-CC (40.8% versus 49.3%, P = 0.059). After adjustment for age, sex, body mass index, diabetes, and excessive alcohol intake, the rs738409 G allele homozygote carriers still carried a higher risk for HS (OR: 1.93, 95% CI: 1.35-2.77, P = 0.003). CONCLUSION: The PNPLA3 rs738409 GG genotype is positively associated with HS, while the IFNL3 rs 12979860 CC genotype may be negatively associated with HS, in Asian CHC patients.


Asunto(s)
Hígado Graso/genética , Hepatitis C Crónica/genética , Interleucinas/genética , Lipasa/genética , Proteínas de la Membrana/genética , Adulto , Anciano , Alelos , Pueblo Asiatico/genética , Hígado Graso/etiología , Hígado Graso/patología , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Hepatitis C Crónica/complicaciones , Hepatitis C Crónica/patología , Humanos , Interferones , Hígado/patología , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple
6.
Int J Mol Sci ; 18(7)2017 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-28644386

RESUMEN

Capsaicin is a potential chemotherapeutic agent for different human cancers. In Southeast China, nasopharyngeal carcinoma (NPC) has the highest incidence of all cancers, but final treatment outcomes are unsatisfactory. However, there is a lack of information regarding the anticancer activity of capsaicin in NPC cells, and its effects on the signaling transduction pathways related to apoptosis and autophagy remain unclear. In the present study, the precise mechanisms by which capsaicin exerts anti-proliferative effects, cell cycle arrest, autophagy and apoptosis were investigated in NPC-TW01 cells. Exposure to capsaicin inhibited cancer cell growth and increased G1 phase cell cycle arrest. Western blotting and quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) were used to measure capsaicin-induced autophagy via involvement of the class III PI3K/Beclin-1/Bcl-2 signaling pathway. Capsaicin induced autophagy by increasing levels of the autophagy markers LC3-II and Atg5, enhancing p62 and Fap-1 degradation and increasing caspase-3 activity to induce apoptosis, suggesting a correlation of blocking the PI3K/Akt/mTOR pathway with the above-mentioned anticancer activities. Taken together, these data confirm that capsaicin inhibited the growth of human NPC cells and induced autophagy, supporting its potential as a therapeutic agent for cancer.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Capsaicina/farmacología , Carcinoma/tratamiento farmacológico , Neoplasias Nasofaríngeas/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Carcinoma/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Humanos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/metabolismo , Nasofaringe/efectos de los fármacos , Nasofaringe/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
7.
Cancer Biol Ther ; 17(10): 1070-1078, 2016 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-27636029

RESUMEN

ROGDI is a protein that contains a leucine zipper domain and may be involved in cell proliferation. In addition, ROGDI is associated with genome stability by regulating the activity of a DNA damage marker, γ-H2AX. The role of ROGDI in tumor radiosensitization has not been investigated. Previous studies have indicated that radiosensitivity is associated with DNA repair and the cell cycle. In general, the G2/M DNA damage checkpoint is more sensitive to radiation, whereas the G1/S phase transition is more resistant to radiation. Inhibition of cyclin-dependent kinases (CDKs) can lead to a halt of cell cycle progression and a stay at different phases or checkpoints. Our data show that the downregulation of ROGDI led to a decreased expression of CDK 1, 2, cyclin A, B and resulted in a G2/M phase transition block. In addition, the downregulation of ROGDI increased cell accumulation at the G2 phase as detected using flow cytometry and decreased cell survival as revealed by clonogenic assay in HeLa and C33A cells following irradiation. These findings suggest that the downregulation of ROGDI can mediate radiosensitivity by blocking cells at G2/M, the most radiosensitive phase of the cell cycle, as well as exerting deleterious effects in the form of DNA damage, as shown by increased γ-H2AX activation.

8.
Sci Rep ; 6: 33816, 2016 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-27665934

RESUMEN

It has been demonstrated that microRNA-122 (miR-122) plays key roles in the modulation of hepatitis B virus (HBV) replication. This study examined the role of miR-122 in patients with hepatitis C virus (HCV)-HBV dual infection with active hepatitis C who received pegylated interferon-α and ribavirin dual therapy. We enrolled 121 patients with HCV-HBV dual infection after dual therapy. Stored serum was collected before treatment. RT-PCR was used to analyze miR-122. HBsAg seroclearance was noted in 37 (30.1%) cases during a median follow-up period of 5.4 years. miR-122 was significantly lower in HBsAg seroclearance patients than in non-HBsAg seroclearance patients (P < 0.014). Multivariate analysis showed that miR-122 was an independent factor of HBsAg seroclearance (OR: 0.30, 95% CI: 0.09-0.98, P = 0.046). miR-122 was significantly higher in patients who were qHBsAg > 100 IU/mL versus ≤100 IU/mL (P < 0.001). We concluded that in patients with HBV-HCV dual infection with active hepatitis C, miR-122 was associated with HBsAg seroclearance after therapy and qHBsAg level before therapy, indicating that miR-122 plays key roles in modulating HBV replication.

9.
Biomed Res Int ; 2016: 3639868, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27595100

RESUMEN

This study investigated the effect of cilostazol on proangiogenesis functions in human early endothelial progenitor cells (EPCs) in vitro and the therapeutic implication of hybrid therapy with cilostazol and human early EPCs in vivo. Cilostazol significantly increased colony-forming units and enhanced differentiation of EPCs toward endothelial lineage. Treatments resulted in antiapoptotic effects and stimulated proliferation and migration and in vitro vascular tube formation through activation of stromal cell-derived factor-1 (SDF-1)/C-X-C chemokine receptor type 4 (CXCR4)/phosphatidylinositol-3 kinase (PI3K)/Akt signaling pathway. Blood flow recovery and capillary density in murine ischemic hindlimbs were significantly improved in cilostazol-treated, human early EPCs-treated, and cotreatment groups. The effects were attenuated with SDF-1α inhibition. Plasma SDF-1α levels were significantly higher in 3 active treatment groups after surgery, with greatest effects observed in hybrid therapy. The angiogenic effects of transplanted EPCs pretreated with cilostazol ex vivo were superior to untreated EPCs using in vivo Matrigel assay. Implanted EPCs were incorporated into the capillary, with pretreatment or cotreatment with cilostazol resulting in enhanced effects. Taken together, cilostazol promotes a large number of proangiogenic functions in human early EPCs through activation of SDF-1/CXCR4/PI3K/Akt signaling, and hybrid therapy provides a synergistic effect in vivo. Cotreatment may be beneficial in ischemic disease.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Quimiocina CXCL12/metabolismo , Células Progenitoras Endoteliales/efectos de los fármacos , Tetrazoles/farmacología , Células Cultivadas , Cilostazol , Sinergismo Farmacológico , Humanos
10.
J Vasc Surg ; 63(4): 1051-62.e3, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25595409

RESUMEN

OBJECTIVE: Cilostazol is an antiplatelet agent with vasodilatory effects that works by increasing intracellular concentrations of cyclic adenosine monophosphate (cAMP). This study investigated the effects of cilostazol in preventing high glucose (HG)-induced impaired angiogenesis and examined the potential mechanisms involving activation of AMP-activated protein kinase (AMPK). METHODS: Assays for colony formation, adhesion, proliferation, migration, and vascular tube formation were used to determine the effect of cilostazol in HG-treated endothelial progenitor cells (EPCs) or human umbilical vein endothelial cells (HUVECs). Animal-based assays were performed in hyperglycemic ICR mice undergoing hind limb ischemia. An immnunoblotting assay was used to identify the expression and activation of signaling molecules in vitro and in vivo. RESULTS: Cilostazol treatment significantly restored endothelial function in EPCs and HUVECs through activation of AMPK/acetyl-coenzyme A carboxylase (ACC)-dependent pathways and cAMP/protein kinase A (PKA)-dependent pathways. Recovery of blood flow in the ischemic hind limb and the population of circulating CD34(+) cells were significantly improved in cilostazol-treated mice, and these effects were abolished by local AMPK knockdown. Cilostazol increased the phosphorylation of AMPK/ACC and Akt/endothelial nitric oxide synthase signaling molecules in parallel with or downstream of the cAMP/PKA-dependent signaling pathway in vitro and in vivo. CONCLUSIONS: Cilostazol prevents HG-induced endothelial dysfunction in EPCs and HUVECs and enhances angiogenesis in hyperglycemic mice by interactions with a broad signaling network, including activation of AMPK/ACC and probably cAMP/PKA pathways.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Inductores de la Angiogénesis/farmacología , Diabetes Mellitus Experimental/enzimología , Células Progenitoras Endoteliales/efectos de los fármacos , Glucosa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Isquemia/tratamiento farmacológico , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/efectos de los fármacos , Tetrazoles/farmacología , Proteínas Quinasas Activadas por AMP/genética , Animales , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cilostazol , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Relación Dosis-Respuesta a Droga , Células Progenitoras Endoteliales/enzimología , Células Progenitoras Endoteliales/patología , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Isquemia/enzimología , Isquemia/patología , Isquemia/fisiopatología , Masculino , Ratones Endogámicos ICR , Fosforilación , Interferencia de ARN , Recuperación de la Función , Flujo Sanguíneo Regional , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección
11.
Cell Cycle ; 14(17): 2764-76, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26114227

RESUMEN

We previously reported that Aurora-A and the hNinein binding protein AIBp facilitate centrosomal structure maintenance and contribute to spindle formation. Here, we report that AIBp also interacts with Plk1, raising the possibility of functional similarity to Bora, which subsequently promotes Aurora-A-mediated Plk1 activation at Thr210 as well as Aurora-A activation at Thr288. In kinase assays, AIBp acts not only as a substrate but also as a positive regulator of both Aurora-A and Plk1. However, AIBp functions as a negative regulator to block phosphorylation of hNinein mediated by Aurora-A and Plk1. These findings suggest a novel AIBp-dependent regulatory machinery that controls mitotic entry. Additionally, knockdown of hNinein caused failure of AIBp to target the centrosome, whereas depletion of AIBp did not affect the localization of hNinein and microtubule nucleation. Notably, knockdown of AIBp in HeLa cells impaired both Aurora-A and Plk1 kinase, resulting in phenotypes with multiple spindle pole formation and chromosome misalignment. Our data show that depletion of AIBp results in the mis-localization of TACC3 and ch-TOG, but not CEP192 and CEP215, suggesting that loss of AIBp dominantly affects the Aurora-A substrate to cause mitotic aberrations. Collectively, our data demonstrate that AIBp contributes to mitotic entry and bipolar spindle assembly and may partially control localization, phosphorylation, and activation of both Aurora-A and Plk1 via hNinein during mitotic progression.


Asunto(s)
Aurora Quinasa A/metabolismo , Proteínas Portadoras/fisiología , Proteínas de Ciclo Celular/metabolismo , Mitosis/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Huso Acromático/metabolismo , Aurora Quinasa A/genética , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN , Células HEK293 , Células HeLa , Humanos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Huso Acromático/genética , Quinasa Tipo Polo 1
12.
Molecules ; 20(6): 11508-23, 2015 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-26111179

RESUMEN

Pluchea indica is used in traditional medicine for the treatment of lumbago, ulcer, tuberculosis and inflammation. The anti-cancer activities and the underlying molecular mechanisms of the ethanolic extracts of P. indica root (PIRE) were characterized in the present study. PIRE strongly inhibited the viability of the human nasopharyngeal carcinoma cells (NPC-TW 01 and NPC-TW 04) in a time- and dose-dependent manner. Migration of cancer cells was also suppressed by PIRE. In addition, PIRE significantly increased the occurrence of the cells in sub-G1 phase and the extent of DNA fragmentation in a dose-dependent manner, which indicates that PIRE significantly increased apoptosis in NPC cells. The apoptotic process triggered by PIRE involved up-regulation of pro-apoptotic Bax protein and down-regulation of anti-apoptotic Bcl-2 protein, consequently increasing the ratios of Bax/Bcl-2 protein levels. Moreover, the p53 protein was up-regulated by PIRE in a concentration-dependent manner. Therefore, PIRE could induce the apoptosis-signaling pathway in NPC cells by activation of p53 and by regulation of apoptosis-related proteins.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Nasofaríngeas/tratamiento farmacológico , Extractos Vegetales/administración & dosificación , Asteraceae/química , Carcinoma , Línea Celular Tumoral , Humanos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/patología , Extractos Vegetales/química
13.
Biochim Biophys Acta ; 1853(8): 1796-807, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25920809

RESUMEN

GSK3ß binding of GSKIP affects neurite outgrowth, but the physiological significance of PKA binding to GSKIP remains to be determined. We hypothesized that GSKIP and GSK3ß mediate cAMP/PKA/Drp1 axis signaling and modulate mitochondrial morphology by forming a working complex comprising PKA/GSKIP/GSK3ß/Drp1. We demonstrated that GSKIP wild-type overexpression increased phosphorylation of Drp1 S637 by 7-8-fold compared to PKA kinase-inactive mutants (V41/L45) and a GSK3ß binding-defective mutant (L130) under H2O2 and forskolin challenge in HEK293 cells, indicating that not only V41/L45, but also L130 may be involved in Drp1-associated protection of GSKIP. Interestingly, silencing either GSKIP or GSK3ß but not GSK3α resulted in a dramatic decrease in Drp1 S637 phosphorylation, revealing that both GSKIP and GSK3ß are required in this novel PKA/GSKIP/GSK3ß/Drp1 complex. Moreover, overexpressed kinase-dead GSK3ß-K85R, which retains the capacity to bind GSKIP, but not K85M which shows total loss of GSKIP-binding, has a higher Drp1 S637 phosphorylation similar to the GSKIP wt overexpression group, indicating that GSK3ß recruits Drp1 by anchoring rather than in a kinase role. With further overexpression of either V41/L45P or the L130P GSKIP mutant, the elongated mitochondrial phenotype was lost; however, ectopically expressed Drp1 S637D, a phosphomimetic mutant, but not S637A, a non-phosphorylated mutant, restored the elongated mitochondrial morphology, indicating that Drp1 is a downstream effector of direct PKA signaling and possibly has an indirect GSKIP function involved in the cAMP/PKA/Drp1 signaling axis. Collectively, our data revealed that both GSKIP and GSK3ß function as anchoring proteins in the cAMP/PKA/Drp1 signaling axis modulating Drp1 phosphorylation.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , GTP Fosfohidrolasas/metabolismo , Glucógeno Sintasa Quinasa 3/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Represoras/fisiología , Células Cultivadas , Dinaminas , GTP Fosfohidrolasas/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Células HEK293 , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias/genética , Dinámicas Mitocondriales/fisiología , Proteínas Mitocondriales/genética , Fosforilación , Proteínas Represoras/metabolismo , Transducción de Señal/genética
14.
Molecules ; 19(11): 17464-77, 2014 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-25356567

RESUMEN

Cancer stem cells (CSCs) are a subset of cancer cells in tumors or established cancer cell lines that can initiate and sustain the growth of tumors in vivo. Cancer stem cells can be enriched in serum-free, suspended cultures that allow the formation of tumorspheres over several days to weeks. Brefeldin A (BFA) is a mycotoxin that induces endoplasmic reticulum (ER) stress in eukaryotic cells. We found that BFA, at sub-microgram per milliliter concentrations, preferentially induced cell death in MDA-MB-231 suspension cultures (EC50: 0.016 µg/mL) compared to adhesion cultures. BFA also effectively inhibited clonogenic activity and the migration and matrix metalloproteinases-9 (MMP-9) activity of MDA-MB-231 cells. Western blotting analysis indicated that the effects of BFA may be mediated by the down-regulation of breast CSC marker CD44 and anti-apoptotic proteins Bcl-2 and Mcl-1, as well as the reversal of epithelial-mesenchymal transition. Furthermore, BFA also displayed selective cytotoxicity toward suspended MDA-MB-468 cells, and suppressed tumorsphere formation in T47D and MDA-MB-453 cells, suggesting that BFA may be effective against breast cancer cells of various phenotypes.


Asunto(s)
Neoplasias de la Mama/patología , Brefeldino A/farmacología , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Técnicas de Cultivo de Célula/métodos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Receptores de Hialuranos/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Micotoxinas/farmacología , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fenotipo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
15.
Int J Mol Sci ; 15(9): 16611-27, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25244013

RESUMEN

Angiogenesis, the process of neovascularization, plays an important role in physiological and pathological conditions. ST104P is a soluble polysulfated-cyclo-tetrachromotropylene compound with anti-viral and anti-thrombotic activities. However, the functions of ST104P in angiogenesis have never been explored. In this study, we investigated the effects of ST104P in angiogenesis in vitro and in vivo. Application of ST104P potently suppressed the microvessels sprouting in aortic rings ex vivo. Furthermore, ST104P treatment significantly disrupted the vessels' development in transgenic zebrafish in vivo. Above all, repeated administration of ST104P resulted in delayed tumor growth and prolonged the life span of mice bearing Lewis lung carcinoma. Mechanistic studies revealed that ST104P potently inhibited the migration, tube formation and wound closure of human umbilical endothelial cells (HUVECs). Moreover, ST104P treatment inhibited the secretion and expression of matrix metalloproteinase-2 (MMP-2) in a dose-dependent manner. Together, these results suggest that ST104P is a potent angiogenesis inhibitor and may hold potential for treatment of diseases due to excessive angiogenesis including cancer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Endotelio Vascular/efectos de los fármacos , Compuestos Macrocíclicos/farmacología , Metaloproteinasa 2 de la Matriz/biosíntesis , Naftalenosulfonatos/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/uso terapéutico , Inhibidores de la Angiogénesis/toxicidad , Animales , Animales Modificados Genéticamente , Aorta , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/efectos de los fármacos , Endotelio Vascular/enzimología , Endotelio Vascular/metabolismo , Inducción Enzimática/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/uso terapéutico , Compuestos Macrocíclicos/toxicidad , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Morfogénesis/efectos de los fármacos , Naftalenosulfonatos/química , Naftalenosulfonatos/uso terapéutico , Naftalenosulfonatos/toxicidad , Neovascularización Patológica/tratamiento farmacológico , Pez Cebra/embriología
16.
Biomed Res Int ; 2014: 910260, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25013810

RESUMEN

Traumatic brain injury (TBI) leads to important and deleterious neuroinflammation, as evidenced by indicators such as edema, cytokine production, induction of nitric oxide synthase, and leukocyte infiltration. After TBI, cerebral vascular endothelial cells play a crucial role in the pathogenesis of inflammation. In our previous study, we proved that simvastatin could attenuate cerebral vascular endothelial inflammatory response in a rat traumatic brain injury. This purpose of this study was to determine whether simvastatin combined with an antioxidant could produce the same effect or greater and to examine affected surrogate biomarkers for the neuroinflammation after traumatic brain injury in rat. In our study, cortical contusions were induced, and the effect of acute and continuous treatment of simvastatin and vitamin C on behavior and inflammation in adult rats following experimental TBI was evaluated. The results demonstrated that simvastatin combined with an antioxidant could provide neuroprotection and it may be attributed to a dampening of cerebral vascular endothelial inflammatory response.


Asunto(s)
Antioxidantes/administración & dosificación , Lesiones Encefálicas/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Simvastatina/administración & dosificación , Animales , Lesiones Encefálicas/patología , Venas Cerebrales/efectos de los fármacos , Venas Cerebrales/lesiones , Combinación de Medicamentos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Humanos , Inflamación/patología , Óxido Nítrico Sintasa/metabolismo , Ratas
17.
Ann Clin Lab Sci ; 44(2): 145-50, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24795052

RESUMEN

AIM: Traumatic brain injury (TBI) leads to important and deleterious inflammation, as evidenced by edema, cytokine production, induction of nitric oxide synthase, and leukocyte infiltration. After TBI, the activation of cerebral vascular endothelial cells plays a crucial role in the pathogenesis of inflammation. In this study, we hypothesized that the activation of cerebral vascular endothelial cells plays a crucial role in the pathogenesis of inflammation and outcome after TBI. It may represent a key cellular target for statin therapy. METHODS: In our study, cortical contusions were induced, and the effect of continuous treatment of simvastatin on behavior and inflammation in adult rats following experimental TBI was evaluated. The treatment group received 15 mg/kg of simvastatin daily for 3 days. Neurological function was assessed with the grip test. RESULTS: The results showed that the non-treatment control group had a significantly greater increase in ICAM-1 expression from pre-injury to the post-injury 72 h time point as compared to the expression in treatment group. The treatment group had better neurological function as evidenced in a grip test performed from baseline to 72 h. The analysis of a western blot test and pathology also demonstrated reduced ICAM-1 expression and a smaller area of damage and tissue loss. CONCLUSION: Our findings suggest that simvastatin could attenuate the activation of cerebral vascular endothelial inflammatory response and decrease the loss of neurological function and brain tissue.


Asunto(s)
Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/tratamiento farmacológico , Cerebro/irrigación sanguínea , Endotelio Vascular/patología , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Simvastatina/uso terapéutico , Animales , Western Blotting , Cerebro/efectos de los fármacos , Cerebro/patología , Endotelio Vascular/efectos de los fármacos , Inmunohistoquímica , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Ratas Sprague-Dawley , Simvastatina/farmacología , Coloración y Etiquetado , Factores de Tiempo
18.
Kaohsiung J Med Sci ; 30(5): 219-23, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24751383

RESUMEN

Mammalian oligosaccharyltransferase complex subunit OSTC/DC2 protein has recently been shown to be a new subunit of the oligosaccharyltransferase; however, its physiological role is still unclear. Here, we report the expression pattern of OSTC/DC2 protein in the context of heat shock stress. Its upregulation was detected both in cells treated with heat shock in vitro and in an animal model of heat shock in vivo. Northern blot analysis indicated that OSTC/DC2 mRNA is ubiquitously expressed in various human tissues, with abundant expression in the placenta and liver. The temporal changes of OSTC/DC2 protein expression following acute heat shock in human malignant glioblastoma cell line U87MG and mice were analyzed by Western blot assay. In general, expression of OSTC/DC2 protein was elevated after heat shock; however, the time courses of the change of OSTC/DC2 protein expression varied in different tissues. In the cerebellum, heat shock induction of OSTC/DC2 protein and activation of AKT, a key regulator of stress response, followed a similar time course. These results suggest that the upregulation of OSTC/DC2, a novel component of the oligosaccharyltransferase complex, is part of the mammalian heat shock response.


Asunto(s)
Respuesta al Choque Térmico , Hexosiltransferasas/metabolismo , Proteínas de la Membrana/metabolismo , Subunidades de Proteína/metabolismo , Animales , Línea Celular Tumoral , Cerebelo/enzimología , Perfilación de la Expresión Génica , Respuesta al Choque Térmico/genética , Hexosiltransferasas/genética , Humanos , Proteínas de la Membrana/genética , Ratones Endogámicos BALB C , Fosforilación , Subunidades de Proteína/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
20.
Kaohsiung J Med Sci ; 29(9): 478-83, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24018150

RESUMEN

Although an increasing number of RNA adenosine-to-inosine (A-to-I) editing sites are being discovered, how the editing frequencies of these sites are modulated to fine-tune protein function in adaptive responses is not well understood. A previous study screening for heat tolerance in Drosophila mutants discovered a hypnos-2 mutant strain that was later found to be defective in dADAR, the Drosophila gene encoding the A-to-I editing enzyme. This supports the hypothesis that cells and organisms respond to stressful environments by ADAR (adenosine deaminase acting on RNA)-mediated RNA editing. Here, we investigated changes in the RNA A-to-I editing frequencies of 30 Drosophila nervous system targets in response to heat shock, a stress acclimatization that requires the dADAR function. To our surprise, most of these nervous system editing targets showed reduced editing. Our results suggest that a change in RNA editing pattern is a mechanism by which organisms acclimate to drastic environmental change. However, how RNA editing confers heat resistance is more complicated and requires further investigation.


Asunto(s)
Adaptación Fisiológica , Adenosina/genética , Drosophila/genética , Calor , Inosina/genética , Edición de ARN , Animales , Drosophila/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...