Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
J Cell Physiol ; 234(5): 6263-6273, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30246389

RESUMEN

Transforming growth factor-ß (TGF-ß) signaling pathway is involved in fibrosis in most, if not all forms of cardiac diseases. Here, we evaluate a positive feedback signaling the loop of TGF-ß1/promyelocytic leukemia (PML) SUMOylation/Pin1 promoting the cardiac fibrosis. To test this hypothesis, the mice underwent transverse aortic constriction (3 weeks) were developed and the morphological evidence showed obvious interstitial fibrosis with TGF-ß1, Pin1 upregulation, and increase in PML SUMOylation. In neonatal mouse cardiac fibroblasts (NMCFs), we found that exogenous TGF-ß1 induced the upregulation of TGF-ß1 itself in a time- and dose-dependent manner, and also triggered the PML SUMOylation and the formation of PML nuclear bodies (PML-NBs), and consequently recruited Pin1 into nuclear to colocalize with PML. Pharmacological inhibition of TGF-ß signal or Pin1 with LY364947 (3 µM) or Juglone (3 µM), the TGF-ß1-induced PML SUMOylation was reduced significantly with downregulation of the messenger RNA and protein for TGF-ß1 and Pin1. To verify the cellular function of PML by means of gain- or loss-of-function, the positive feedback signaling loop was enhanced or declined, meanwhile, TGF-ß-Smad signaling pathway was activated or weakened, respectively. In summary, we uncovered a novel reciprocal loop of TGF-ß1/PML SUMOylation/Pin1 leading to myocardial fibrosis.


Asunto(s)
Miocardio/patología , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Proteína de la Leucemia Promielocítica/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Retroalimentación Fisiológica , Fibrosis , Corazón , Cardiopatías/metabolismo , Cardiopatías/patología , Ratones , Sumoilación
2.
J Cell Physiol ; 233(12): 9575-9583, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29943817

RESUMEN

The epithelial-mesenchymal transition (EMT) is a key event associated with metastasis and dissemination in breast tumor pathogenesis. Promyelocytic leukemia (PML) gene produces several isoforms due to alternative splicing; however, the biological function of each specific isoform has yet to be identified. In this study, we report a previously unknown role for PMLIV, the most intensely studied nuclear isoform, in transforming growth factor-ß (TGF-ß) signaling-associated EMT and migration in breast cancer. This study demonstrates that PMLIV overexpression promotes a more aggressive mesenchymal phenotype and increases the migration of MCF-7 cancer cells. This event is associated with activation of the TGF-ß canonical signaling pathway through the induction of Smad2/3 phosphorylation and the translocation of phospho-Smad2/3 to the nucleus. In this study, we report a previously unknown role for PMLIV in TGF-ß signaling-induced regulation of breast cancer-associated EMT and migration. Targeting this pathway may be therapeutically beneficial.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular , Transición Epitelial-Mesenquimal , Proteína de la Leucemia Promielocítica/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Núcleo Celular/metabolismo , Femenino , Células HEK293 , Humanos , Células MCF-7 , Modelos Biológicos , Fosforilación , Proteína de la Leucemia Promielocítica/química , Dominios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Transducción de Señal , Proteína Smad2/metabolismo , Proteína smad3/metabolismo
3.
Toxicol Appl Pharmacol ; 345: 1-9, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29524504

RESUMEN

BACKGROUND AND PURPOSE: Protein modification by small ubiquitin-like modifier (SUMO) plays a critical role in the pathogenesis of heart diseases. The present study was designed to determine whether ginkgolic acid (GA) as a SUMO-1 inhibitor exerts an inhibitory effect on cardiac fibrosis induced by myocardial infarction (MI). EXPERIMENTAL APPROACH: GA was delivered by osmotic pumps in MI mice. Masson staining, electron microscopy (EM) and echocardiography were used to assess cardiac fibrosis, ultrastructure and function. Expression of SUMO-1, PML, TGF-ß1 and Pin1 was measured with Western blot or Real-time PCR. Collagen content, cell viability and myofibroblast transformation were measured in neonatal mouse cardiac fibroblasts (NMCFs). Promyelocytic leukemia (PML) protein was over-expressed by plasmid transfection. KEY RESULTS: GA improved cardiac fibrosis and dysfunction, and decreased SUMO-1 expression in MI mice. GA (>20 µM) inhibited NMCF viability in a dose-dependent manner. Nontoxic GA (10 µM) restrained angiotensin II (Ang II)-induced myofibroblast transformation and collagen production. GA also inhibited expression of TGF-ß1 mRNA and protein in vitro and in vivo. GA suppressed PML SUMOylation and PML nuclear body (PML-NB) organization, and disrupted expression and recruitment of Pin1 (a positive regulator of TGF-ß1 mRNA), whereas over-expression of PML reversed that. CONCLUSIONS AND IMPLICATIONS: Inhibition of SUMO-1 by GA alleviated MI-induced heart dysfunction and fibrosis, and the SUMOylated PML/Pin1/TGF-ß1 pathway is crucial for GA-inhibited cardiac fibrosis.


Asunto(s)
Infarto del Miocardio/tratamiento farmacológico , Proteína SUMO-1/antagonistas & inhibidores , Salicilatos/uso terapéutico , Animales , Animales Recién Nacidos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Relación Dosis-Respuesta a Droga , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Fibrosis/patología , Masculino , Ratones , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Proteína SUMO-1/metabolismo , Salicilatos/farmacología , Volumen Sistólico/efectos de los fármacos , Volumen Sistólico/fisiología
4.
Mol Ther ; 25(3): 666-678, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28143738

RESUMEN

The promyelocytic leukemia protein (PML) is essential in the assembly of dynamic subnuclear structures called PML nuclear bodies (PML-NBs), which are involved in regulating diverse cellular functions. However, the possibility of PML being involved in cardiac disease has not been examined. In mice undergoing transverse aortic constriction (TAC) and arsenic trioxide (ATO) injection, transforming growth factor ß1 (TGF-ß1) was upregulated along with dynamic alteration of PML SUMOylation. In cultured neonatal mouse cardiac fibroblasts (NMCFs), ATO, angiotensin II (Ang II), and fetal bovine serum (FBS) significantly triggered PML SUMOylation and the assembly of PML-NBs. Inhibition of SUMOylated PML by silencing UBC9, the unique SUMO E2-conjugating enzyme, reduced the development of cardiac fibrosis and partially improved cardiac function in TAC mice. In contrast, enhancing SUMOylated PML accumulation, by silencing RNF4, a poly-SUMO-specific E3 ubiquitin ligase, accelerated the induction of cardiac fibrosis and promoted cardiac function injury. PML colocalized with Pin1 (a positive regulator for TGF-ß1 mRNA expression in PML-NBs) and increased TGF-ß1 activity. These findings suggest that the UBC9/PML/RNF4 axis plays a critical role as an important SUMO pathway in cardiac fibrosis. Modulating the protein levels of the pathway provides an attractive therapeutic target for the treatment of cardiac fibrosis and heart failure.


Asunto(s)
Silenciador del Gen , Miocardio/metabolismo , Miocardio/patología , Proteínas Nucleares/genética , Proteína de la Leucemia Promielocítica/metabolismo , Factores de Transcripción/genética , Enzimas Ubiquitina-Conjugadoras/genética , Angiotensina II/farmacología , Animales , Trióxido de Arsénico , Arsenicales/farmacología , Colágeno/biosíntesis , Fibrosis , Ratones , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Óxidos/farmacología , Unión Proteica , Sumoilación , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Ubiquitina-Proteína Ligasas
5.
Cell Physiol Biochem ; 41(2): 835-848, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28214884

RESUMEN

BACKGROUND/AIMS: The present study investigated whether the transient receptor potential melastatin 4 (TRPM4) channel plays a role in high salt diet (HSD)-induced endothelial injuries. METHODS: Western blotting and immunofluorescence were used to examine TRPM4 expression in the mesenteric endothelium of Dahl salt-sensitive (SS) rats fed a HSD. The MTT, TUNEL, and transwell assays were used to evaluate the cell viability, cell apoptosis, and cell migration, respectively, of human umbilical vein endothelial cells (HUVECs). Enzyme-linked immunosorbent assays were used to determine the concentrations of intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion protein 1 (VCAM-1), and E-selectin. Carboxy-H2DCFDA, a membrane-permeable reactive oxygen species (ROS)-sensitive fluorescent probe, was used to detect intracellular ROS levels. RESULTS: TRPM4 was mainly expressed near the plasma membrane of mesenteric artery endothelial cells, and its expression level increased in SS hypertensive rats fed a HSD. Its protein expression was significantly upregulated upon treatment with exogenous hydrogen peroxide (H2O2) and aldosterone in cultured HUVECs. Cell viability decreased upon treatment with both agents in a concentration-dependent manner, which could be partially reversed by 9-phenanthrol, a specific TRPM4 inhibitor. Exogenous H2O2 induced apoptosis, enhanced cell migration, and increased the release of adhesion molecules, including ICAM-1, VCAM-1, and E-selectin, all of which were significantly attenuated upon treatment with 9-phenanthrol. Aldosterone and H2O2 induced the accumulation of intracellular ROS, which was significantly inhibited by 9-phenanthrol, suggesting that oxidative stress is one of the mechanisms underlying aldosterone-induced endothelial injury. CONCLUSIONS: Given the fact that oxidative stress and high levels of circulating aldosterone are present in hypertensive patients, we suggest that the upregulation of TRPM4 in the vascular endothelium may be involved in endothelial injuries caused by these stimuli.


Asunto(s)
Dieta , Endotelio Vascular/metabolismo , Canales Catiónicos TRPM/metabolismo , Aldosterona/toxicidad , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Selectina E/análisis , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Peróxido de Hidrógeno/toxicidad , Masculino , Arterias Mesentéricas/citología , Estrés Oxidativo/efectos de los fármacos , Fenantrenos/farmacología , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Endogámicas Dahl , Cloruro de Sodio/farmacología , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/genética , Regulación hacia Arriba/efectos de los fármacos
6.
Front Pharmacol ; 11: 123, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32153415

RESUMEN

Arsenic trioxide (ATO) is an effective therapeutic agent against acute promyelocytic leukemia (APL); however, its anti-tumor effect on solid tumors such as colorectal cancer (CRC) is still in debate. Ascorbic acid (AA) also produces a selective cytotoxic activity against tumor cells. Here, we exploit the potential benefit of ATO/AA combination in generating cytotoxicity to CRC cells, which may lay the groundwork for the potential combinational chemotherapy of CRCs. According to the results, we found that ATO and AA effectively inhibited the viability of human CRC cells in a synergistic manner. AA and ATO corporately activated caspase-3 to trigger apoptosis and upregulated the expression of caspase-1 and promoted formation of inflammasomes to induce pyroptosis. Furthermore, the stimulation of reactive oxygen species (ROS) overproduction was demonstrated as a subcellular mechanism for apoptosis and pyroptosis induced by ATO/AA combination treatment. Our findings suggest that ATO combination with a conventional dosage of AA offers an advantage for killing CRC cells. The synergistic action of ATO/AA combination might be considered a plausible strategy for the treatment of CRC and perhaps other solid tumors as well.

7.
Mol Ther Oncolytics ; 16: 86-99, 2020 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-31970286

RESUMEN

Small ubiquitin-related modifiers (SUMO) represent a class of ubiquitin-like proteins that are conjugated, like ubiquitin, by a set of enzymes to form cellular regulatory proteins, and play key roles in the control of cell proliferation, differentiation, and apoptosis. We found that ginkgolic acid (GA) can significantly reduce cell vitality in a dose- and time-dependent manner and can also accelerate cyto-apoptosis in both Tca8113 and Cal-27 cells. Migration and wound-healing assays were executed to determine the anti-migration effect of GA in oral squamous cell carcinoma (OSCC) cell lines. GA represses transforming growth factor-ß1 (TGF-ß1)-induced epithelial-mesenchymal transition (EMT) markers in OSCC cell lines. This investigation is the first evidence that GA suppresses TGF-ß1-induced SUMOylation of SMAD4. We show that GA affects the phosphorylation of SMAD2/3 protein and the release of SMAD4. In the xenograft mouse model, the OSCC progression was reduced by GA, effectively suppressing the growth of tumors. In addition, siSMAD4 improved cell migration and viability, which was inhibited by GA in Tca8113 cells. GA suppresses tumorigenicity and tumor progression of OSCC through inhibition of TGF-ß1-induced enhancement of SUMOylation of SMAD4. Thus, GA could be a promising therapeutic for OSCC.

8.
Br J Pharmacol ; 175(15): 3111-3130, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29722931

RESUMEN

BACKGROUND AND PURPOSE: PEP06, a polypeptide modified from endostatin, was investigated for its antitumour effects on colorectal cancer (CRC) and the possible mechanisms of this antitumour activity were examined in in vitro and in vivo models. EXPERIMENTAL APPROACH: After PEP06 treatment, cell proliferation and migration assays were performed in CRC cells. Epithelial-mesenchymal transition (EMT) progression was determined by Western blotting, immunofluorescent staining and immunohistochemistry in vitro and in a residual xenograft model. MiRNAs regulated by PEP06 were identified by miRNA microarray and verified by in situ hybridization and quantitative real-time PCR. The interactions between PEP06 and integrin αvß3 were determined with Biacore SA biochips. The cellular function of miR-146b-5p was validated by gain-of-function and loss-of-function approaches. A mouse model of lung metastasis was used to determine the effect of PEP06 on metastatic growth. KEY RESULTS: PEP06 did not affect cell viability but reduced migration and EMT in SW620 and HCT116 cells. PEP06 significantly repressed the expression of miR-146b-5p in these two cell lines through binding to integrin αvß3. MiR-146b-5p was shown to increase EMT by targeting Smad4, and the miR-146b-5p-Smad4 cascade regulated EMT in CRC. PEP06 also suppressed CRC pulmonary metastasis, increased survival of mice and hampered residual tumour growth by inhibiting EMT through down-regulating miR-146b-5p. CONCLUSIONS AND IMPLICATIONS: PEP06 is a polypeptide that inhibits the growth and metastasis of colon cancer through its RGD motif binding to integrin αvß3, thereby down-regulating miR-146b-5p to inhibit EMT in vitro and in vivo. It might have potential as a therapeutic for CRC.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Animales , Antígenos CD/metabolismo , Antineoplásicos/farmacología , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Endostatinas , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Neoplasias Pulmonares/secundario , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/metabolismo , Fragmentos de Péptidos , Vimentina/metabolismo , Cicatrización de Heridas/efectos de los fármacos
9.
Oncotarget ; 8(28): 45447-45458, 2017 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-28525371

RESUMEN

The human ether-a-go-go-related gene (HERG) channel is a novel target for the treatment of drug-induced long QT syndrome, which causes lethal cardiotoxicity. This study is designed to explore the possible role of PML SUMOylation and its associated nuclear bodies (NBs) in the regulation of HERG protein expression. Both arsenic trioxide (ATO) and angiotensin II (Ang II) were able to significantly reduce HERG protein expression, while also increasing PML SUMOylation and accelerating the formation of PML-NBs. Pre-exposure of cardiomyocytes to a SUMOylation chemical inhibitor, ginkgolic acid, or the silencing of UBC9 suppressed PML SUMOylation, subsequently preventing the downregulation of HERG induced by ATO or Ang II. Conversely, knockdown of RNF4 led to a remarkable increase in PML SUMOylation and the function of PML-NBs, further promoting ATO- or Ang II-induced HERG protein downregulation. Mechanistically, an increase in PML SUMOylation by ATO or Ang II dramatically enhanced the formation of PML and Pin1 complexes in PML-NBs, leading to the upregulation of TGF-ß1 protein, eventually inhibiting HERG expression through activation of protein kinase A. The present work uncovered a novel molecular mechanism underlying HERG protein expression and indicated that PML SUMOylation is a critical step in the development of drug-acquired arrhythmia.


Asunto(s)
Angiotensina II/farmacología , Arsenicales/farmacología , Canal de Potasio ERG1/metabolismo , Óxidos/farmacología , Animales , Trióxido de Arsénico , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Canal de Potasio ERG1/genética , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Cuerpos de Inclusión Intranucleares/metabolismo , Ratones , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Proteína de la Leucemia Promielocítica/metabolismo , Sumoilación/efectos de los fármacos , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo
10.
J Am Heart Assoc ; 6(6)2017 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-28559372

RESUMEN

BACKGROUND: Myocardial infarction (MI) is often accompanied by cardiomyocyte apoptosis, which decreases heart function and leads to an increased risk of heart failure. The aim of this study was to examine the effects of transforming growth factor-ß receptor III (TGFßR3) on cardiomyocyte apoptosis during MI. METHODS AND RESULTS: An MI mouse model was established by left anterior descending coronary artery ligation. Cell viability, apoptosis, TGFßR3, and mitogen-activated protein kinase signaling were assessed by methylthiazolyldiphenyl-tetrazolium bromide assay, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay, immunofluorescence, electron microscopy, and Western blotting. Our results demonstrated that TGFßR3 expression in the border region of the heart was dynamically changed during MI. After stimulation with H2O2, TGFßR3 overexpression in cardiomyocytes led to increased cell apoptosis and activation of p38 signaling, whereas TGFßR3 knockdown had the opposite effect. ERK1/2 and JNK1/2 signaling was not altered by TGFßR3 modulation, and p38 inhibitor (SB203580) reduced the effect of TGFßR3 on apoptosis, suggesting that p38 has a nonredundant function in activating apoptosis. Consistent with the in vitro observations, cardiac TGFßR3 transgenic mice showed augmented cardiomyocyte apoptosis, enlarged infarct size, increased injury, and enhanced p38 signaling upon MI. Conversely, cardiac loss of function of TGFßR3 by adeno-associated viral vector serotype 9-TGFßR3 short hairpin RNA attenuated the effects of MI in mice. CONCLUSIONS: TGFßR3 promotes apoptosis of cardiomyocytes via a p38 pathway-associated mechanism, and loss of TGFßR3 reduces MI injury, which suggests that TGFßR3 may serve as a novel therapeutic target for MI.


Asunto(s)
Apoptosis , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Proteoglicanos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Peróxido de Hidrógeno/farmacología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Proteoglicanos/genética , Interferencia de ARN , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal , Factores de Tiempo , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Basic Clin Pharmacol Toxicol ; 98(1): 104-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16433899

RESUMEN

We used Fluo-3/AM to examine the effect of platelet-activating factor on the intracellular Ca2+([Ca2+]i) levels in isolated myocytes of guinea pig ventricle. Myocytes were isolated with Langendorff perfusion technique and were challenged with platelet-activating factor. Addition of platelet-activating factor (1 pM to 10 nM) significantly increased the [Ca2+]i in the presence and absence of extracellular Ca2+. The notion that increases in intracellular Ca2+ induced by platelet-activating factor is the result of stimulation of intracellular Ca2+ pool rather than increasing Ca2+ influx was further supported by the whole cell patch-clamp experiments in which the platelet-activating factor did not alter the activity of L-type of Ca2+ channels (I(Ca-L)). Treatment of myocytes with ryanodine failed to abolish the stimulatory effect of platelet-activating factor on [Ca2+]i. In contrast, inhibition of IP3-sensitive Ca2+ release pool with 2-aminoethoxydiphenyl borate (2-APB) blocked the effect of platelet-activating factor. We conclude that the platelet-activating factor-induced increase in intracellular Ca2+ is mediated by stimulation of IP3 receptor but not by stimulation of I(Ca-L) and ryanodine-sensitive receptor.


Asunto(s)
Canales de Calcio/efectos de los fármacos , Señalización del Calcio , Miocitos Cardíacos/efectos de los fármacos , Factor de Activación Plaquetaria/farmacología , Receptores Citoplasmáticos y Nucleares/efectos de los fármacos , Compuestos de Anilina , Animales , Compuestos de Boro/farmacología , Calcio/metabolismo , Canales de Calcio/metabolismo , Relación Dosis-Respuesta a Droga , Colorantes Fluorescentes , Cobayas , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Técnicas In Vitro , Receptores de Inositol 1,4,5-Trifosfato , Potenciales de la Membrana , Miocitos Cardíacos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Tiempo , Xantenos
12.
Basic Clin Pharmacol Toxicol ; 98(4): 381-8, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16623862

RESUMEN

Arsenic trioxide (As(2)O(3)) has been found to be effective for relapsed or refractory acute promyelocytic leukaemia, but its clinical use is burdened by QT prolongation, Torsade de pointes tachycardias, and sudden cardiac death. The aim of the present study was to elucidate the ionic mechanisms of As(2)O(3)-induced abnormalities of cardiac electrophysiology and the therapeutic action of choline on As(2)O(3)-caused QT prolongation in guinea pig. Intravenous administration of As(2)O(3) prolonged the QT interval in a dose- and time-dependent manner in guinea pig hearts, and the QT prolongation could be modulated by choline. By using whole-cell patch clamp technique and confocal laser scanning microscopy, we found that As(2)O(3) significantly lengthened action potential duration measured at 50 and 90% of repolarization, enhanced L-type calcium currents (I(Ca-L)), inhibited delayed rectifier potassium currents (I(K)), and increased intracellular calcium concentration ([Ca(2+)](i)) in guinea pig ventricular myocytes. Choline corrected As(2)O(3)-mediated alterations of action potential duration, I(Ca-L) and [Ca(2+)](i), but had no effect on the I(K) inhibition. As(2)O(3) markedly disturbed the normal equilibrium of transmembrane currents (increasing I(Ca-L) and suppressing I(K)) in guinea pig cardiomyocyte, and induced prolongation of action potential duration, further degenerated into QT prolongation. Choline normalized QT interval abnormality and corrected lengthened action potential duration by inhibiting the elevated I(Ca-L) and [Ca(2+)](i) in ventricular myocytes during As(2)O(3) application.


Asunto(s)
Arsenicales/farmacología , Colina/farmacología , Corazón/efectos de los fármacos , Síndrome de QT Prolongado/prevención & control , Óxidos/farmacología , Animales , Trióxido de Arsénico , Calcio/fisiología , Electrocardiografía , Femenino , Cobayas , Corazón/fisiología , Técnicas In Vitro , Síndrome de QT Prolongado/inducido químicamente , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp
13.
Zhonghua Yu Fang Yi Xue Za Zhi ; 37(3): 158-60, 2003 May.
Artículo en Zh | MEDLINE | ID: mdl-12880559

RESUMEN

OBJECTIVE: To study the effect of exogenous nucleic acid on physical functions, morphology of hepatic cells and brain neurons in aged rats. METHODS: Thirty two aged Wistar rats (20 month-old) were divided randomly into four groups (one aged control group and three aged experimental groups) and eight young rats (3 month-old) was set as young control group. Control groups were fed on standard chow and experimental groups were fed on standard chow supplemented with 93.75 mg/kg (high-dosage group), 46.88 mg/kg (middle-dosage group) and 9.38 mg/kg (low-dosage group) of yeast RNA respectively. SOD, MDA, HDL, sex hormone and growth hormone were determined at the end of a 4-week observation. The microcosmic images of the hepatic cells and brain neurons using the image-pro plus (V.4.0) were also observed. RESULTS: SOD, serum HDL and growth hormone levels in the high dosage group were significantly higher (P < 0.05) than that in the aged control group, and the levels were not different from that in the young control group. MDA level of all yeast RNA supplemented groups was significantly lower than that of aged control group (P < 0.05) and that was not different from the young control group. Serum testosterone of the high and middle dosage groups reached the level of young control group, and that was much higher than the aged control and low dosage group (P < 0.05). Estradiol levels among the aged rats were not different, and those were much lower than the young control group (P < 0.05). Much more number of brain neurons were observed in the high-dose group than other aged rats (P < 0.05). Brain neurons, hepatic cells and karyons in the high-dose group were bigger than that in other aged rats (P < 0.05). CONCLUSION: Exogenous yeast RNA might play an important role in physical functions, the morphology of brain neurons and hepatic cells in natural aged rats. There might have a dose-effect relationship in the process.


Asunto(s)
Envejecimiento , Encéfalo/fisiología , Hepatocitos/ultraestructura , Hígado/fisiología , ARN de Hongos/farmacología , Levaduras/química , Animales , Encéfalo/ultraestructura , Relación Dosis-Respuesta a Droga , Hígado/ultraestructura , Masculino , Neuronas/ultraestructura , Distribución Aleatoria , Ratas , Ratas Wistar
14.
Zhonghua Yu Fang Yi Xue Za Zhi ; 37(6): 398-402, 2003 Nov.
Artículo en Zh | MEDLINE | ID: mdl-14703491

RESUMEN

OBJECTIVE: To investigate the effect and its mechanism of salmon milt DNA (SMD) on age-related involutions in mouse thymus. METHODS: Female BALB/C mice aged 10 months were divided randomly into three groups according to their weights. They were high dosage group (333.33 mg.kg(-1).b.w.d(-1)), low dosage group (166.67 mg.kg(-1).b.w.d(-1)) and control group (0 mg.kg(-1).b.w.d(-1)). After five weeks, their thymus indexes were measured and the thymocytes were counted and the thymus cortex thicknesses were also measured using Image-Pro Plus (version. 4.0) software in the thymus section. All the data were analyzed by SAS statistic software. Microarray technique was applied to screen the gene fragments, which were differentially expressed between the high dosage group and the control group, together with RT-PCR to further confirm some of them. RESULTS: No significant differences of the variables including body weight, thymus weight and thymus indexes among the three groups were found. The thymocytes quantities of thymus cortex and medulla in the high dosage group were significantly higher than those of the control group (P < 0.01 and P < 0.05, respectively). The thymus cortex thicknesses of both SMD supplement groups were significantly higher than that of the control group (P < 0.01 and P < 0.05 respectively). 112 differently expressed gene fragments were isolated. Furthermore, we found the fragments with the logged number of U23789, X80232 and Aw209102 were highly expressed in the high dosage group when RT-PCR technique was used. CONCLUSIONS: SMD may reverse the age-related involutions in mouse thymus via up-regulation the expression of proliferation related genes and via up-regulation the expression of development and differentiation related genes simultaneously.


Asunto(s)
Envejecimiento/efectos de los fármacos , ADN/administración & dosificación , Expresión Génica/efectos de los fármacos , Salmón/genética , Timo/efectos de los fármacos , Actinas/genética , Envejecimiento/genética , Animales , División Celular/efectos de los fármacos , División Celular/genética , Femenino , Ratones , Ratones Endogámicos BALB C , Proyectos Piloto , Distribución Aleatoria , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Timo/citología , Timo/metabolismo
15.
PLoS One ; 8(8): e73424, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23977387

RESUMEN

OBJECTIVE: Regulation of apical calcium entry is important for the function of principal cells of the collecting duct. However, the molecular identity and the regulators of the transporter/channel, which is responsible for apical calcium entry and what factors regulate the calcium conduction remain unclear. METHODS AND RESULTS: We report that endogenous TRPP2 and TRPV4 assemble to form a 23-pS divalent cation-permeable non-selective ion channel at the apical membrane of renal principal cells of the collecting duct. TRPP2\TRPV4 channel complex was identified by patch-clamp, immunofluorescence and co-immunprecipitation studies in both principal cells that either possess normal cilia (cilia (+)) or in which cilia are absent (cilia (-)). This channel has distinct biophysical and pharmacological and regulatory profiles compared to either TRPP2 or TRPV4 channels. The rate of occurrence detected by patch clamp was higher in cilia (-) compared to cilia (+) cells. In addition, shRNA knockdown of TRPP2 increased the prevalence of TRPV4 channel activity while knockdown of TRPV4 resulted in TRPP2 activity and knockdown of both proteins vastly decreased the 23-pS channel activity. Epidermal growth factor (EGF) stimulated TRPP2\TRPV4 channel through the EGF receptor (EGFR) tyrosine kinase-dependent signaling. With loss of cilia, apical EGF treatment resulted in 64-fold increase in channel activity in cilia (-) but not cilia (+) cells. In addition EGF increased cell proliferation in cilia (-) cell that was dependent upon TRPP2\TRPV4 channel mediated increase in intracellular calcium. CONCLUSION: We conclude that in the absence of cilia, an EGF activated TRPP2\TRPV4 channel may play an important role in increased cell proliferation and cystogenesis.


Asunto(s)
Agonistas de los Canales de Calcio/farmacología , Calcio/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Túbulos Renales Colectores/metabolismo , Canales Catiónicos TRPP/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Cationes Bivalentes/metabolismo , Proliferación Celular/efectos de los fármacos , Cilios/metabolismo , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnica del Anticuerpo Fluorescente , Silenciador del Gen/efectos de los fármacos , Inmunoprecipitación , Activación del Canal Iónico/efectos de los fármacos , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos
16.
Naunyn Schmiedebergs Arch Pharmacol ; 383(3): 321-30, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21279332

RESUMEN

Maslinic acid (MA) is a triterpenoid with a high concentration that exists in olives. This natural compound, which has shown multiple biological activities, was proved to be an anti-tumoral agent more recently. We have investigated the mechanisms of MA with regard to its inhibitory effects on the growth of salivary gland adenoid cystic carcinoma (ACC). We demonstrated that MA at 10-100 µM reduced cell viability in a dose-dependent manner, IC(50) of 43.68 µM, and 45.76 µM, respectively in cultured ACC-2 and ACC-M cells. Fifty micromolars of MA efficiently induced apoptosis as indicated by AO/EB staining, electronic microscopy, flow cytometry, and activation of caspase-3 activity. MA induced an elevation of [Ca(2+)](i) in a dose-dependent manner, and cell viability inhibition and cell apoptosis induced by MA were blocked by an intracellular Ca(2+) chelator, BAPTA-AM. The elevation of [Ca(2+)](i) induced by MA was blocked by EGTA or TRPV channel inhibitor suggesting TRPV channel involved in calcium influx induced by MA. MA also activated ERK and p38 MAPK in a time-dependent manner. MA induced cell apoptosis and activation of caspase-3 activity were reversed by SB203580, but not by PD98059, suggesting that the apoptosis induction of MA was via p38 MAPK, but not via ERK. Chelation of intracellular Ca(2+) with BAPTA reversed MA induced p38 MAPK phosphorylation, but SB203580 did not block MA-evoked elevation of [Ca(2+)](i), suggesting a Ca(2+)-evoked p38 MAPK signaling involved in MA-induced apoptosis in ACC cells. Taken together, in ACC cells, maslinic acid induced an increase in [Ca(2+)](i), which evoked p38 MAPK phosphorylation, subsequently activated caspase-3 leading to apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Señalización del Calcio/fisiología , Carcinoma Adenoide Quístico/patología , Neoplasias de las Glándulas Salivales/patología , Transducción de Señal/fisiología , Triterpenos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Antineoplásicos Fitogénicos/farmacología , Señalización del Calcio/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quelantes/farmacología , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Rojo de Rutenio/farmacología , Transducción de Señal/efectos de los fármacos , Canales Catiónicos TRPC/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
17.
Int J Cardiol ; 143(1): 86-93, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-19237210

RESUMEN

BACKGROUND: Previous studies suggested that platelet-activating factor (PAF) plays an important role in ischemic diseases. Apoptosis has been implicated in myocardial infarction-related cell death. The present study was designed to determine whether PAF could induce apoptosis in cardiac myocytes and the underlying mechanisms by which PAF causes apoptosis. METHODS: H9c2 cardiac myocytes were used to investigate the effect of PAF on intracellular calcium concentration, cell viability and cell apoptosis. Signaling pathway of caspase-3, cytochrome c and MAPK (ERK, JNK, p38) was determined during the PAF induced apoptosis. RESULTS: First, our results showed that treatment of H9c2 cardiomyocytes with PAF (0.2 to 20 microM) caused apoptosis in these cells and the apoptotic process was suppressed by either BN52021 (an antagonist of PAF receptor) or BAPTA/AM (an intracellular Ca2+ chelator), suggesting an involvement of PAF and its receptor mediated calcium-dependent signaling. Second, we found that activity of p38-MAPK (mitogen-activated protein kinase) and caspase-3 was elevated in the cells treated with PAF, without altering activity of ERK and JNK, and that PAF-induced enhancement of caspase-3 activity was attenuated by application of either BAPTA/AM or SB203580 (p38 inhibitor). Furthermore, PAF-induced apoptosis and release of cytochrome c from mitochondria was blunted by SB203580, and PAF-induced enhancement of p38 activity was also attenuated by BAPTA/AM. CONCLUSION: Our data implicate that a PAF and its receptor in triggering apoptosis occurs in cultured H9c2 cardiac myocytes via a calcium-dependent p38 MAPK activated cytochrome c/caspase-3 apoptosis signaling pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Calcio/metabolismo , Miocitos Cardíacos , Factor de Activación Plaquetaria/farmacología , Factor de Activación Plaquetaria/fisiología , Animales , Apoptosis/fisiología , Caspasa 3/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Quelantes/farmacología , Citocromos c/metabolismo , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Oral Oncol ; 45(11): 998-1004, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19589718

RESUMEN

New chemotherapeutic strategy should be investigated to enhance clinical management in salivary gland adenoid cystic carcinoma (ACC). Recently, sulforaphane (SFN), as a natural compound from cruciferous vegetables exhibits a potent anti-cancer activity in various tumor cells, but remains uncertain in ACC cells. The present study examined whether SFN suppresses proliferation and in ACC cells, if so, the possible molecular targets would be further investigated. Cell survives, apoptosis, cell cycle progression and molecular targets were identified by multiple detecting techniques, including trypan blue dye exclusion assay, electron microscopy, AO/EB staining, flow cytometry and immunoblotting in human lung high metastasis cell line of salivary gland adenoid cystic carcinoma (ACC-M). The results showed that 5-20 microM SFN suppressed proliferation and induced apoptosis of ACC-M cells in dose- and time-dependent manners. Cell cycle analysis demonstrated treatment of ACC-M cells with 20 microM SFN resulted in G(2)/M cell cycle arrest, which was associated with a marked decline in protein levels of G(2)/M regulatory proteins including cyclin B1 and cyclin-dependent kinase 1 (Cdk1). In terms of apoptosis, SFN increased the expression of Bax and decreased the level of Bcl-2 and subsequently triggered release of cytochrome c from mitochondria and activation of caspase-3, but Fas level and caspase-8 activity remained unchanged at all time points. Furthermore, levels of nuclear factor-kappaB (NF-kappaB) p65 in both of the cytoplasm and the nucleus have also been markedly suppressed by SFN in a time-dependent manner. Taken together, these results suggest SFN inhibits cell growth via inducing G(2)/M cell arrest and apoptosis in ACC-M cells. These events have been associated with SFN-regulated multiple targets involved in ACC-M cell proliferation. The present study provides an evidence for testing SFN efficacy in vivo and warranting future investigations to exam the clinical potential of SFN in ACC chemotherapy.


Asunto(s)
Anticarcinógenos/farmacología , Carcinoma Adenoide Quístico/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias de las Glándulas Salivales/tratamiento farmacológico , Tiocianatos/farmacología , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Carcinoma Adenoide Quístico/secundario , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Proteínas del Ojo/metabolismo , Humanos , Isotiocianatos , Neoplasias Pulmonares/secundario , Proteínas de la Membrana/metabolismo , Neoplasias de las Glándulas Salivales/patología , Sulfóxidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA