Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Circ J ; 83(4): 718-726, 2019 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-30787218

RESUMEN

BACKGROUND: Intracellular uric acid is known to increase the protein level and channel current of atrial Kv1.5; however, mechanisms of the uric acid-induced enhancement of Kv1.5 expression remain unclear. Methods and Results: The effects of uric acid on mRNA and protein levels of Kv1.5, as well as those of Akt, HSF1 and Hsp70, in HL-1 cardiomyocytes were studied by using qRT-PCR and Western blotting. The uptake of uric acid was measured using radio-labeled uric acid. The Kv1.5-mediated channel current was also measured by using patch clamp techniques. Uric acid up-taken by HL-1 cells significantly increased the level of Kv1.5 proteins in a concentration-dependent manner, with this increase abolished by an uric acid transporter inhibitor. Uric acid slowed degradation of Kv1.5 proteins without altering its mRNA level. Uric acid enhanced phosphorylation of Akt and HSF1, and thereby increased both transcription and translation of Hsp70; these effects were abolished by a PI3K inhibitor. Hsp70 knockdown abolished the uric acid-induced increases of Kv1.5 proteins and channel currents. CONCLUSIONS: Intracellular uric acid could stabilize Kv1.5 proteins through phosphorylation of Akt and HSF1 leading to enhanced expression of Hsp70.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Factores de Transcripción del Choque Térmico/metabolismo , Canal de Potasio Kv1.5/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ácido Úrico/farmacología , Animales , Línea Celular , Canal de Potasio Kv1.5/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos , Biosíntesis de Proteínas , Transcripción Genética
2.
Circ J ; 80(12): 2443-2452, 2016 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-27803431

RESUMEN

BACKGROUND: Long QT syndrome 2 (LQT2) is caused by mutations in the human ether-a-go-go-related gene (hERG). Most of its mutations give rise to unstable hERG proteins degraded by the proteasome. Recently, carbachol was reported to stabilize the wild-type hERG-FLAG via activation of the muscarinic type 3 receptor (M3-mAChR). Its action on mutant hERG-FLAG, however, remains uninvestigated.Methods and Results:A novel mutant hERG-FLAG carried 2 mutations: an amino acid substitution G572S and an in-frame insertion D1037_V1038insGD. When expressed in HEK293 cells, this mutant hERG-FLAG was degraded by the proteasome and failed to be transported to the cell surface. Carbachol restored stability of the mutant hERG-FLAG and facilitated cell-surface expression. Carbachol activated PKC, augmented phosphorylation of heat shock factor 1 (HSF1) and enhanced expression of heat shock proteins (hsps), hsp70 and hsp90. Both a M3-mAChR antagonist, 4-DAMP, and a PKC inhibitor, bisindolylmaleimide, abolished carbachol-induced stabilization of the mutant hERG-FLAG. CONCLUSIONS: M3-mAChR activation leads to enhancement of hsp expression via PKC-dependent phosphorylation of HSF1, thereby stabilizing the mutant hERG-FLAG protein. Thus, M3-mAChR activators may have a therapeutic value for patients with LQT2. (Circ J 2016; 80: 2443-2452).


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Canal de Potasio ERG1 , Síndrome de QT Prolongado , Mutación , Receptor Muscarínico M3/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Adolescente , Proteínas de Unión al ADN/genética , Canal de Potasio ERG1/genética , Canal de Potasio ERG1/metabolismo , Células HEK293 , Factores de Transcripción del Choque Térmico , Humanos , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/metabolismo , Masculino , Fosforilación/genética , Estabilidad Proteica , Receptor Muscarínico M3/genética , Factores de Transcripción/genética , Transfección
3.
Circ J ; 79(12): 2659-68, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26477273

RESUMEN

BACKGROUND: Hyperuricemia induces endothelial dysfunction, oxidative stress and inflammation, increasing cardiovascular morbidities. It also raises the incidence of atrial fibrillation; however, underlying mechanisms are unknown. METHODS AND RESULTS: The effects of urate on expression of Kv1.5 in cultured mouse atrial myocytes (HL-1 cells) using reverse transcriptase-PCR, immunoblots, flow cytometry and patch-clamp experiments were studied. Treatment with urate at 7 mg/dl for 24 h increased the Kv1.5 protein level, enhanced ultra-rapid delayed-rectifier K(+)channel currents and shortened action potential duration in HL-1 cells. HL-1 cells expressed the influx uric acid transporter (UAT), URATv1, and the efflux UATs, ABCG2 and MRP4. An inhibitor against URATv1, benzbromarone, abolished the urate effects, whereas an inhibitor against ABCG2, KO143, augmented them. Flow cytometry showed that urate induced an increase in reactive oxygen species, which was abolished by the antioxidant, N-acetylcysteine (NAC), and the NADPH-oxidase inhibitor, apocynin. Both NAC and apocynin abolished the enhancing effects of urate on Kv1.5 expression. A urate-induced increase in the Kv1.5 proteins was accompanied by phosphorylation of extracellular signal-regulated kinase (ERK), and was abolished by an ERK inhibitor, PD98059. NAC abolished phosphorylation of ERK by urate. CONCLUSIONS: Intracellular urate taken up by UATs enhanced Kv1.5 protein expression and function in HL-1 atrial myocytes, which could be attributable to ERK phosphorylation and oxidative stress derived from nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Hiperuricemia/metabolismo , Canal de Potasio Kv1.5/biosíntesis , Proteínas Musculares/metabolismo , Miocitos Cardíacos/metabolismo , Ácido Úrico/farmacología , Animales , Línea Celular , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Hiperuricemia/patología , Canal de Potasio Kv1.5/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos
4.
Clin Exp Nephrol ; 19(4): 576-84, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25239792

RESUMEN

BACKGROUND: Familial juvenile hyperuricemic nephropathy (FJHN) is an autosomal dominant disorder caused by mutations in UMOD that encodes uromodulin. Topiroxostat, a novel non-purine analog, selectively inhibits xanthine oxidase and reduces the serum uric acid levels and the urinary albuminuria. METHODS: Genomic DNA of a patient was extracted from peripheral white blood. Exons and flanking sequences of UMOD were amplified by PCR with primers. Mutation analysis was performed by direct sequencing of the PCR products. The wild-type and mutant uromodulin were expressed in HEK293 cells and analyzed by western blotting, immunoprecipitation, immunofluorescence, and flow cytometry. RESULTS: We identified an FJHN patient who carried a novel UMOD mutation G335A (C112Y). The levels of both cytosolic and secreted C112Y protein were significantly decreased compared with the wild-type, whereas the level of ubiquitination was higher in C112Y than that in the wild type. The half-life of C112Y was shortened and it was restored by a proteasome inhibitor MG132. Immunofluorescence revealed decreased levels of C112Y in the Golgi apparatus and on the plasma membrane. Expression of C112Y induced cellular apoptosis as revealed by flow cytometry. Apoptosis induced by C112Y was suppressed by topiroxostat. CONCLUSION: C112Y causes its protein instability resulting cellular apoptosis which could be suppressed with topiroxostat.


Asunto(s)
Apoptosis/efectos de los fármacos , Gota/genética , Hiperuricemia/genética , Enfermedades Renales/genética , Nitrilos/uso terapéutico , Piridinas/uso terapéutico , Uromodulina/genética , Adulto , Gota/tratamiento farmacológico , Células HEK293 , Humanos , Hiperuricemia/tratamiento farmacológico , Enfermedades Renales/tratamiento farmacológico , Masculino , Mutación , Nitrilos/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Piridinas/farmacología
5.
Biochem Biophys Res Commun ; 431(2): 225-31, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23313480

RESUMEN

P19 embryonal carcinoma (EC) cells are pluripotent stem cells and have numerous morphological and biochemical properties in common with embryonic stem (ES) cells. However, P19 cells differentiate very ineffectively as embryoid bodies (EBs) without the specific chemical inducers whereas ES cells exhibit spontaneous differentiation to the three germ layers. Recently the heterochromatin protein 1 (HP1) family protein HP1γ, which is an epigenetic modulator that binds histone H3 methylated at lysine 9, is shown to be associated with the progression from pluripotent to differentiated status in ES cells. Therefore, to study the role of HP1γ in the differentiation capacity of P19 cells, we have established a HP1γ-overexpressing P19 cell line (HPlγ-P19). Similar to the parental P19 cells, undifferentiated HP1γ-P19 cells continued to express pluripotency marker genes. However, HP1γ-P19 cells exhibited significant morphological differentiation including beating cardiomyocytes, as well as Tuj1-positive neuronal cells and Sox17-positive endodermal cells after EB formation under a normal culture condition. Moreover, real-time RT-qPCR analysis revealed that HP1γ-P19 EB cells expressed various differentiation marker genes. Thus, HP1γ-P19 cells could give rise to all three germ layers in EBs without any drug treatment. Therefore, HP1γ affects the spontaneous differentiation potential of P19 cells, and might play major roles in the decision of cell fates in pluripotent stem cells.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas Cromosómicas no Histona/biosíntesis , Células Madre de Carcinoma Embrionario/citología , Células Germinativas/citología , Células Madre Pluripotentes/citología , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/genética , Expresión Génica , Humanos
6.
Circ J ; 76(12): 2875-83, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22972200

RESUMEN

BACKGROUND: The prion protein (PrP) has been reported to serve as a surface maker for isolation of cardiomyogenic progenitors from murine embryonic stem (ES) cells. Although PrP-positive cells exhibited automaticity, their electrophysiological characteristics remain unresolved. The aim of the present study was therefore to investigate the electrophysiological properties of PrP-positive cells in comparison with those of HCN4p-or Nkx2.5-positive cells. METHODS AND RESULTS: Differentiation of AB1, HCN5p-EGFP and hcgp7 ES cells into cardiac progenitors was induced by embryoid body (EB) formation. EBs were dissociated and cells expressing PrP, HCN4-EGFP and/or Nkx2.5-GFP were collected via flow cytometry. Sorted cells were subjected to reverse transcriptase-polymerase chain reaction, immunostaining and patch-clamp experiments. PrP-positive cells expressed mRNA of undifferentiation markers, first and second heart field markers, and cardiac-specific genes and ion channels, indicating their commitment to cardiomyogenic progenitors. PrP-positive cells with automaticity showed positive and negative chronotropic responses to isoproterenol and carbamylcholine, respectively. Hyperpolarization-activated cation current (I(f)) was barely detectable, whereas Na(+) and L-type Ca(2+) channel currents were frequently observed. Their spontaneous activity was slowed by inhibition of sarcoplasmic reticulum Ca(2+) uptake and release but not by blocking I(f). The maximum diastolic potential of their spontaneous firings was more depolarized than that of Nkx2.5-GFP-positive cells. CONCLUSIONS: PrP-positive cells contained cardiac progenitors that separated from the lineage of sinoatrial node cells. PrP can be used as a marker to enrich nascent cardiac progenitors.


Asunto(s)
Potenciales de Acción , Células Madre Embrionarias/metabolismo , Miocitos Cardíacos/metabolismo , Priones/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Linaje de la Célula , Separación Celular/métodos , Técnicas de Cocultivo , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Citometría de Flujo , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Ratones , Ratones de la Cepa 129 , Contracción Miocárdica , Técnicas de Placa-Clamp , Periodicidad , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección
7.
Circ Rep ; 2(8): 425-432, 2020 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-33693264

RESUMEN

Background: Monocarboxylate transporter 9 (MCT9), an orphan transporter member of the solute carrier family 16 (SLC16), possibly reabsorbs uric acid in the renal tubule and has been suggested by genome-wide association studies to be involved in the development of hyperuricemia and gout. In this study we investigated the mechanisms regulating the expression of human (h) MCT9, its degradation, and physiological functions. Methods and Results: hMCT9-FLAG was stably expressed in HEK293 cells and its degradation, intracellular localization, and urate uptake activities were assessed by pulse-chase analysis, immunofluorescence, and [14C]-urate uptake experiments, respectively. hMCT9-FLAG was localized on the plasma membrane as well as in the endoplasmic reticulum and Golgi apparatus. The proteasome inhibitors MG132 and lactacystine increased levels of hMCT9-FLAG protein expression with enhanced ubiquitination, prolonged their half-life, and decreased [14C]-urate uptake. [14C]-urate uptake was increased by both heat shock (HS) and the HS protein inducer geranylgeranylacetone (GGA). Both HS and GGA restored the [14C]-urate uptake impaired by MG132. Conclusions: hMCT9 does transport urate and is degraded by a proteasome, inhibition of which reduces hMCT9 expression on the cell membrane and urate uptake. HS enhanced urate uptake through hMCT9.

8.
Dev Growth Differ ; 51(2): 81-93, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19207180

RESUMEN

Embryonic carcinoma (EC) cells, which are malignant stem cells of teratocarcinoma, have numerous morphological and biochemical properties in common with pluripotent stem cells such as embryonic stem (ES) cells. However, three EC cell lines (F9, P19 and PCC3) show different developmental potential and self-renewal capacity from those of ES cells. All three EC cell lines maintain self-renewal capacity in serum containing medium without Leukemia Inhibitory factor (LIF) or feeder layer, and show limited differentiation capacity into restricted lineage and cell types. To reveal the underlying mechanism of these characteristics, we took the approach of characterizing extrinsic factors derived from EC cells on the self-renewal capacity and pluripotency of mouse ES cells. Here we demonstrate that EC cell lines F9 and P19 produce factor(s) maintaining the undifferentiated state of mouse ES cells via an unidentified signal pathway, while P19 and PCC3 cells produce self-renewal factors of ES cells other than LIF that were able to activate the STAT3 signal; however, inhibition of STAT3 activation with Janus kinase inhibitor shows only partial impairment on the maintenance of the undifferentiated state of ES cells. Thus, these factors present in EC cells-derived conditioned medium may be responsible for the self-renewal capacity of EC and ES cells independently of LIF signaling.


Asunto(s)
Factores Biológicos/farmacología , Células Madre de Carcinoma Embrionario/metabolismo , Células Madre Pluripotentes/efectos de los fármacos , Transducción de Señal , Animales , Factores Biológicos/metabolismo , Biomarcadores , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Medios de Cultivo/farmacología , Medios de Cultivo Condicionados/farmacología , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes Reporteros , Estratos Germinativos/citología , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/fisiología , Factor Inhibidor de Leucemia/fisiología , Ratones , Células Madre Pluripotentes/citología , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/fisiología
9.
Biomed Res ; 38(4): 229-238, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28794400

RESUMEN

Proepicardium (PE) cells generate cardiac fibroblasts, smooth muscle cells (SMCs) and endothelial cells that form coronary arteries. T-box18 (Tbx18) is a well-known marker of PE cells and epicardium. We examined whether Tbx18-positive cells differentiated from murine embryonic stem (ES) cells serve as PE progenitors to give rise to vascular SMCs and fibroblasts. To collect Tbx18-positive cells, we established Tbx18-EGFP knock-in mouse ES cells using the CRISPR/Cas9 system. We harvested the Tbx18-EGFP-positive cells on day 8, 10 and 14 after the initiation of differentiation; Tbx18 mRNA was enriched on day 8 to 14 and Snai2 mRNA was enriched on day 8 and 10, indicating successful collection of Tbx18-positive cells. Tbx18-EGFP-positive cells expressed the PE marker WT1 on day 8 and 10. They also expressed the SMC marker Acta2 and fibroblast markers Thy1 and Fsp1 on day 8 to 14, but did not express the endothelial cell marker PECAM or the cardiac cell marker CD166 or Myh7. In conclusion, Tbx18-positive cells represent a part of PE cells in the initial phase of differentiation and subsequently include SMCs as well as fibroblasts. These results indicate that Tbx18-positive cells serve as a PE progenitor to supply a variety of cells that contribute to the formation of coronary arteries.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Fibroblastos/citología , Fibroblastos/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Pericardio/citología , Proteínas de Dominio T Box/metabolismo , Animales , Biomarcadores , Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Técnica del Anticuerpo Fluorescente , Expresión Génica , Técnicas de Sustitución del Gen , Orden Génico , Marcación de Gen , Genes Reporteros , Vectores Genéticos/genética , Ratones , Microscopía Fluorescente , Músculo Liso Vascular/citología , Pericardio/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas de Dominio T Box/genética
10.
Yonago Acta Med ; 59(2): 126-34, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27493483

RESUMEN

BACKGROUND: The prion protein (PrP) might be useful as a tool to collect cardiac progenitor cells derived from embryonic stem (ES) cells. It is also possible that PrP(+) cells include undifferentiated cells with a capacity to develop into tumors. METHODS: PrP(+) cells isolated from embryoid bodies (EB) formed by mouse AB1 ES cells were examined using RT-PCR analysis and clonogeneic cell assay. To assess their potential to differentiate into cardiomyocytes, Nkx2.5(GFP/+) (hcgp7) cells, another ES cell line that carries the GFP reporter gene in the Nkx2.5 loci, were used. RESULTS: PrP(+) cells isolated from EB of day 7 and 14 did not express pluripotency markers, but expressed cardiac cell markers, while PrP(+) cells isolated from EB of day 21 expressed pluripotency markers. Cultured PrP(+) cells isolated from EB of day 21 expressed pluripotency markers to form colonies, whereas those isolated from EB of day 7 and 14 did not. To exclude proliferating cells from PrP(+) cells, stage specific embryo antigen 1 (SSEA1) was employed as a second marker. PrP(+)/SSEA1(-) cells did not proliferate and expressed cardiac cell markers, while PrP(+)/SSEA1(+) did proliferate. CONCLUSION: PrP(+) cells isolated from EB included undifferentiated cells in day 21. PrP(+)/SSEA1(-) cells included cardiomyoctes, suggesting PrP and SSEA1 may be useful as markers to enrich the fraction of cardiomyocytes.

11.
Yonago Acta Med ; 56(4): 93-5, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24574578

RESUMEN

Besides its antiarrhythmic effect on atrial fibrillation, bepridil protects tissue, yet its effect on apoptosis has never been fully tested. We examine the effect of bepridil on apoptosis of HL-1 cells expressing E334K myosin-binding protein C (MyBPC), a model cell of apoptosis. Bepridil was compared with amiodarone, and its effects on the expression of pro- and anti-apoptotic protein and apoptosis of HL-1 cells expressing mutant E334K MyBPC-green fluorescent protein (GFP) was analyzed using Western blot and a flow cytometer. Bepridil decreased the protein levels of both Bax and cytochrome c of cells expressing E334K MyBPC-GFP with no changes in p53 and Bcl-2, while amiodarone decreased cytochrome c but did not influence Bax except in its highest concentration. It also decreased the number of Annexin-V positive cells of HL-1 cells expressing E334K MyBPC-GFP, and decreased apoptosis of HL-1 cells expressing E334K MyBPC-GFP.

12.
Eur J Pharmacol ; 696(1-3): 28-34, 2012 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-23026372

RESUMEN

While bepridil has been reported to alter the stability of ion channel proteins, the precise mechanism of action remains unclear. We examined the effect of bepridil on the stability of Kv1.5 channel proteins expressed in COS7 cells. Bepridil at 0.3-30 µM increased the protein level of Kv1.5 channels in a concentration-dependent manner. Chase experiments showed that bepridil delayed the degradation process of Kv1.5 channel proteins in the same manner as a proteasomal inhibitor, MG132, did. Bepridil increased the immunofluorescent signal of Kv1.5 channel proteins in the endoplasmic reticulum (ER) and Golgi apparatus and on the cell surface. The cell fraction experiment also showed bepridil-induced increases in Kv1.5 in the ER, Golgi apparatus, and the cell membrane. Bepridil at a lower concentration of 1 µM had no effect on the proteasome activity in vitro. A blocker of the ultrarapid delayed-rectifier K(+) channel current, 4-aminopyridine (4AP), abolished bepridil-induced increases in Kv1.5. Kv1.5-medicated membrane currents measured as 4AP-sensitive currents were increased by bepridil. Taken together, we conclude that bepridil stabilizes Kv1.5 proteins at the ER through an action as a chemical chaperone, thereby increasing the density of Kv1.5 channels in the cell membrane.


Asunto(s)
Bepridil/farmacología , Canal de Potasio Kv1.5/metabolismo , Moduladores del Transporte de Membrana/farmacología , 4-Aminopiridina/farmacología , Animales , Células COS , Chlorocebus aethiops , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA