Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Pineal Res ; 68(3): e12631, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31943334

RESUMEN

In the elderly with atherosclerosis, hypertension and diabetes, vascular calcification and ageing are ubiquitous. Melatonin (MT) has been demonstrated to impact the cardiovascular system. In this study, we have shown that MT alleviates vascular calcification and ageing, and the underlying mechanism involved. We found that both osteogenic differentiation and senescence of vascular smooth muscle cells (VSMCs) were attenuated by MT in a MT membrane receptor-dependent manner. Moreover, exosomes isolated from VSMCs or calcifying vascular smooth muscle cells (CVSMCs) treated with MT could be uptaken by VSMCs and attenuated the osteogenic differentiation and senescence of VSMCs or CVSMCs, respectively. Moreover, we used conditional medium from MT-treated VSMCs and Transwell assay to confirm exosomes secreted by MT-treated VSMCs attenuated the osteogenic differentiation and senescence of VSMCs through paracrine mechanism. We also found exosomal miR-204/miR-211 mediated the paracrine effect of exosomes secreted by VSMCs. A potential target of these two miRs was revealed to be BMP2. Furthermore, treatment of MT alleviated vascular calcification and ageing in 5/6-nephrectomy plus high-phosphate diet-treated (5/6 NTP) mice, while these effects were partially reversed by GW4869. Exosomes derived from MT-treated VSMCs were internalised into mouse artery detected by in vivo fluorescence image, and these exosomes reduced vascular calcification and ageing of 5/6 NTP mice, but both effects were largely abolished by inhibition of exosomal miR-204 or miR-211. In summary, our present study revealed that exosomes from MT-treated VSMCs could attenuate vascular calcification and ageing in a paracrine manner through an exosomal miR-204/miR-211.


Asunto(s)
Melatonina/farmacología , MicroARNs/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Calcificación Vascular/metabolismo , Envejecimiento , Animales , Diferenciación Celular/efectos de los fármacos , Exosomas/química , Exosomas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Calcificación Vascular/fisiopatología
2.
Toxicol Appl Pharmacol ; 272(3): 591-7, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23921150

RESUMEN

Ghrelin is a 28-amino-acid peptide that acts as a natural endogenous ligand of the growth hormone secretagogue receptor (GHSR) and strongly stimulates the release of growth hormone from the hypothalamus-pituitary axis. Previous studies have identified the important physiological effects of ghrelin on bone metabolism, such as regulating proliferation and differentiation of osteoblasts, independent of GH/IGF-1 axis. However, research on effects and mechanisms of ghrelin on osteoblast apoptosis is still rare. In this study, we identified expression of GHSR in MC3T3-E1 cells and determined the effects of ghrelin on the apoptosis of osteoblastic MC3T3-E1 cells and the mechanism involved. Our data demonstrated that ghrelin inhibited the apoptosis of osteoblastic MC3T3-E1 cells induced by serum deprivation, as determined by terminal deoxynucleotidyl transferase-mediated deoxyribonucleotide triphosphate nick end-labeling (TUNEL) and ELISA assays. Moreover, ghrelin upregulated Bcl-2 expression and downregulated Bax expression in a dose-dependent manner. Our study also showed decreased activated caspase-3 activity under the treatment of ghrelin. Further study suggested that ghrelin stimulated the phosphorylation of ERK and AKT. Pretreatment of cells with the ERK inhibitor PD98059, PI3K inhibitor LY294002, and GHSR-siRNA blocked the ghrelin-induced activation of ERK and AKT, respectively; however, ghrelin did not stimulate the phosphorylation of p38 or JNK. PD90859, LY294002 and GHSR-siRNA attenuated the anti-apoptosis effect of ghrelin in MC3T3-E1 cells. In conclusion, ghrelin inhibits the apoptosis of osteoblastic MC3T3-E1 cells induced by serum deprivation, which may be mediated by activating the GHSR/ERK and GHSR/PI3K/AKT signaling pathways.


Asunto(s)
Apoptosis/fisiología , Ghrelina/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Osteoblastos/fisiología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Células 3T3 , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Datos de Secuencia Molecular , Osteoblastos/efectos de los fármacos , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología
3.
Amino Acids ; 44(3): 961-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23135225

RESUMEN

It has been hypothesized that adipocytokines originating from adipose tissue may have an important role in bone metabolism. Vaspin is a novel adipocytokine isolated from visceral white adipose tissue, which has been reported to have anti-apoptotic effects in vascular endothelial cells. However, to the best of our knowledge there is no information regarding the effects of vaspin on osteoblast apoptosis. This study therefore examined the possible effects of vaspin on apoptosis in human osteoblasts (hOBs). Our study established that vaspin inhibits hOBs apoptosis induced by serum deprivation, as determined by ELISA and TUNEL assays. Western blot analysis revealed that vaspin upregulates the expression of Bcl-2 and downregulates that of Bax in a dose-dependent manner. Vaspin stimulated the phosphorylation of ERK, and pretreatment of hOBs with the ERK inhibitor PD98059 blocked the vaspin-induced activation of ERK, however, vaspin did not stimulate the phosphorylation of p38, JNK or Akt. Vaspin protects hOBs from serum deprivation-induced apoptosis, which may be mediated by activating the MAPK/ERK signaling pathway.


Asunto(s)
Apoptosis , Sistema de Señalización de MAP Quinasas , Osteoblastos/citología , Serpinas/metabolismo , Células Cultivadas , Humanos , Osteoblastos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
4.
Amino Acids ; 43(5): 2125-36, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22532031

RESUMEN

Apelin receptor (APJ) deficiency has been reported to be preventive against atherosclerosis. However, the mechanism of this effect remains unknown. In this study, quantitative real-time RT-PCR, Western blotting and ELISA analyses revealed a significant increase in the expression of intercellular adhesion molecule-1(ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and monocyte chemoattractant protein-1 (MCP-1) in human umbilical vein endothelial cells (HUVECs) treated with apelin. Inhibitors of cellular signal transduction molecules were used to demonstrate involvement of nuclear factor kappa-B (NF-κB) and c-Jun N-terminal kinase (JNK) pathways in apelin-APJ-induced activation of adhesion molecules and chemokines. Inhibition of APJ expression by RNA interference abrogated apelin-induced expression of adhesion molecules and chemokines and apelin-stimulated cellular signal transduction in HUVECs. The apelin-APJ system in endothelial cells is involved in the expression of adhesion molecules and chemokines, which are important for the initiation of endothelial inflammation-related atherosclerosis. Therefore, apelin-APJ and the cell signaling pathways activated by this system in endothelial cells may represent targets for therapy of atherosclerosis.


Asunto(s)
Quimiocina CCL2/genética , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/farmacología , Receptores Acoplados a Proteínas G/genética , Molécula 1 de Adhesión Celular Vascular/genética , Apelina , Receptores de Apelina , Western Blotting , Células Cultivadas , Quimiocina CCL2/metabolismo , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/metabolismo
5.
Amino Acids ; 39(1): 89-99, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19915792

RESUMEN

Several studies have suggested a direct link between taurine and bone homeostasis. However, the mechanisms of taurine on the regulation of bone metabolism have not been elucidated. Using a coculture of osteoblasts and bone marrow cells as a model for the study of osteoclastogenesis, RANKL-stimulated RAW264.7 cells and M-CSF- and RANKL-induced bone marrow macrophages were investigated to elucidate the possible roles of taurine in osteoclastogenesis. Taurine inhibited osteoclastogenesis in the coculture of osteoblasts and bone marrow cells, but did not influence the expression of OPG and RANKL in osteoblasts. The taurine transporter (TAUT) expressed by RAW264.7 and bone marrow macrophages exhibited typical taurine uptake activity. Taurine directly reduced osteoclastogenesis in RANKL-stimulated RAW264.7 cells and M-CSF- and RANKL-induced bone marrow macrophages, while TAUT siRNA relieved this effect. Our study demonstrated that taurine directly inhibited osteoclastogenesis through the taurine transporter. Taken together, these data suggest that taurine plays a direct role in bone homeostasis by inhibiting osteoclastogenesis.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Taurina/farmacología , Animales , Células de la Médula Ósea/metabolismo , Técnicas de Cocultivo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos ICR , Osteoclastos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Amino Acids ; 39(5): 1193-200, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20495838

RESUMEN

Apoptosis of vascular smooth muscle cells (VSMCs) plays an important role in regulating vascular remodeling during cardiovascular diseases. Apelin is the endogenous ligand for the G-protein-coupled receptor APJ and plays an important role in the cardiovascular system. However, the mechanisms of apelin on apoptosis of VSMCs have not been elucidated. Using a culture of human VSMCs as a model for the study of apoptosis, the relationship between apelin and apoptosis of human VSMCs and the signal pathway involved were investigated. Using western blotting, we confirmed that VSMCs could express APJ. To evaluate the possible role of apelin in VSMC apoptosis, we assessed its effect on apoptosis of human VSMCs. The results showed that apelin inhibited human VSMCs apoptosis induced by serum deprivation. Suppression of APJ with small-interfering RNA (siRNA) abolished the anti-apoptotic activity of apelin. Apelin increased Bcl-2 protein expression, but decreased Bax protein expression. An increase in activation of extracellular signal-regulated protein kinase (ERK) and Akt (a downstream effector of phosphatidylinositol 3-kinase) was shown after apelin stimulation. Suppression of APJ with siRNA abolished the apelin-induced activation of ERK and Akt. LY294002 (a PI3-K inhibitor) blocked apelin-induced activation of Akt and abolished the apelin-induced antiapoptotic activity. Our study suggests that apelin suppresses serum deprivation-induced apoptosis of human VSMCs, and that the anti-apoptotic action is mediated through the APJ/PI3-K/Akt signaling pathways.


Asunto(s)
Apoptosis , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Músculo Liso Vascular/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Apelina , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Humanos , Transducción de Señal/efectos de los fármacos
7.
Aging (Albany NY) ; 11(14): 5232-5245, 2019 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-31352437

RESUMEN

Cortisol-producing adenoma (CPA) is the main cause of Adrenal Cushing syndrome. However, its molecular mechanism is not fully understood. Previous study revealed Synaptophysin (SYP) is ubiquitously expressed in adrenocortical tumors, but its function in CPA still need to be discovered. In the present study we determine the molecular mechanism involved in SYP dysregulation in CPA and how SYP affects the secretion of cortisol in CPA. Our results showed that aberrant DNA methylation of SYP is involved in CPA progress. Using a miRNA microarray and qRT-PCR, we found decreased expression of miR-27a-5p in CPA compared with normal adrenal tissue. Moreover, the expression of TET3, the target gene of miR-27a-5p, increased in CPA compared with normal adrenal tissue. Knock-down of TET3 resulted in hypermethylation of SYP which reducing the expression level of SYP in H295R cells. The miR-27a-5p-TET3-SYP signalling pathway may regulate proliferation and cortisol secretion in H295R cells and, thus, play a key role in CPA development.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/metabolismo , Adenoma Corticosuprarrenal/genética , Adenoma Corticosuprarrenal/metabolismo , Metilación de ADN , Hidrocortisona/metabolismo , Sinaptofisina/genética , Adulto , Línea Celular Tumoral , Dioxigenasas/genética , Femenino , Técnicas de Silenciamiento del Gen , Marcación de Gen , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Transducción de Señal , Regulación hacia Arriba
8.
Endocr Relat Cancer ; 26(5): 525-538, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30870812

RESUMEN

Tumour-derived exosomes under hypoxic conditions contain informative miRNAs involved in the interaction of cancer and para-carcinoma cells, thus contributing to tissue remodelling of the tumour microenvironment (TME). Exosomes isolated from hypoxic papillary thyroid cancer cells, BCPAP cells and KTC-1 cells enhanced the angiogenesis of human umbilical vein endothelial cells (HUVECs) compared with exosomes isolated from normal thyroid follicular cell line (Nthy-ori-3-1), normoxic BCPAP or KTC-1 cells both in vitro and in vivo. miR-21-5p was significantly upregulated in exosomes from papillary thyroid cancer BCPAP cells under hypoxic conditions, while the exosomes isolated from hypoxic BCPAP cells with knockdown of miR-21-5p attenuated the promoting effect of angiogenesis. In addition, our experiment revealed that miR-21-5p directly targeted and suppressed TGFBI and COL4A1, thereby increasing endothelial tube formation. Furthermore, elevated levels of exosomal miR-21-5p are found in the sera of papillary thyroid cancer patients, which promote the angiogenesis of HUVECs. Taken together, our study reveals the cell interaction between hypoxic papillary thyroid cancer cells and endothelial cells, elucidating a new mechanism by which hypoxic papillary thyroid cancer cells increase angiogenesis via exosomal miR-21-5p/TGFBI and miR-21-5p/COL4A1 regulatory pathway.


Asunto(s)
Biomarcadores de Tumor/sangre , Exosomas/metabolismo , MicroARNs/sangre , Neovascularización Patológica/patología , Cáncer Papilar Tiroideo/irrigación sanguínea , Neoplasias de la Tiroides/irrigación sanguínea , Microambiente Tumoral , Animales , Estudios de Casos y Controles , Proliferación Celular , Colágeno Tipo IV/sangre , Proteínas de la Matriz Extracelular/sangre , Regulación Neoplásica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Hipoxia , Masculino , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Pronóstico , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/metabolismo , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Factor de Crecimiento Transformador beta/sangre , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Mol Med Rep ; 19(5): 3807-3814, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30896834

RESUMEN

MicroRNAs (miRNAs) are novel key regulators of cellular differentiation. miR­124 has been reported to regulate osteogenic differentiation of bone marrow­derived mesenchymal stem cells (BMSCs). However, the specific mechanisms involved have not yet been fully elucidated. The present study aimed to investigate the effect of miR­124 on osteogenic differentiation of BMSCs and its underlying mechanisms. In the present study, it was found that alkaline phosphatase (ALP) activity, osteocalcin (OC) secretion, and the protein levels of osterix (Sp7) and runt­related transcription factor 2 (Runx2) were significantly increased, whereas the expression of miR­124 was decreased in a time­dependent manner during osteogenic differentiation of BMSCs. Following overexpression of miR­124 via transfection of miR­124 mimics in BMSCs, Runx2 protein expression and ALP activity were significantly decreased. By contrast, inhibition of miR­124 expression led to an increase in ALP activity and Runx2 expression. Sp7 expression was suppressed in BMSCs transfected with miR­124 mimics while increased when miR­124 expression was inhibited, indicating that miR­124 regulates the expression of Sp7. Moreover, a luciferase reporter assay further verified that Sp7 is the direct target of miR­124. Finally, the effect of miR­124 inhibitor on promoting the differentiation of BMSCs was abolished following treatment with a small interfering RNA targeting Sp7. Taken together, the present study demonstrates that miR­124 inhibits the osteogenic differentiation of BMSCs by targeting Sp7.


Asunto(s)
Diferenciación Celular , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/citología , MicroARNs/genética , Osteogénesis , Factor de Transcripción Sp7/metabolismo , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/metabolismo , Factor de Transcripción Sp7/genética
10.
Aging (Albany NY) ; 11(10): 3182-3197, 2019 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-31129659

RESUMEN

Vascular calcification is one of the most important factors for cardiovascular and all-cause mortality in patients with end-stage renal diseases (ESRD). The current study was aimed to investigate the function and mechanisms of miR-34b on the calcification of vascular smooth muscle cells (VSMCs) both in vitro and in vivo. We found that the expression of miR-34b was significantly suppressed in VSMCs with high inorganic phosphate (Pi) treatment, as well as mouse arteries derived from 5/6 nephrectomy with a high-phosphate diet (0.9% Pi, 5/6 NTP) and human renal arteries from uraemia patients. Overexpression of miR-34b alleviated calcification of VSMCs, while VSMCs calcification was enhanced by inhibiting the expression of miR-34b. Bisulphite sequencing PCR (BSP) uncovered that CpG sites upstream of miR-34b DNA were hypermethylated in calcified VSMCs and calcified arteries due to 5/6 NTP, as well as calcified renal arterial tissues from uraemia patients. Meantime, increased DNA methyltransferase 3a (DNMT3a) resulted in the hypermethylation of miR-34b in VSMCs, while 5-aza-2'-deoxycytidine (5-aza) reduced the methylation rate of miR-34b and restored the expression of miR-34b in VSMCs. When DNMT3a was knocked down using DNMT3a siRNA, the effect of 3.5 mM of Pi on calcification of VSMCs was abrogated. In addition, Notch1 was validated as the functional target of miR-34b and involved in the process of calcification of VSMCs. Taken together, our data showed a specific role for miR-34b in regulating calcification of VSMCs both in vitro and in vivo, which was regulated by upstream DNA methylation of miR-34b and modulated by the downstream target gene expression, Notch1. These results suggested that modulation of miR-34b may offer new insight into a novel therapeutic approach for vascular calcification.


Asunto(s)
Metilación de ADN , MicroARNs/metabolismo , Receptor Notch1/metabolismo , Calcificación Vascular/metabolismo , Animales , Diferenciación Celular , Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Humanos , Masculino , Ratones Endogámicos C57BL , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Osteoblastos , Arteria Renal/metabolismo , Uremia/metabolismo
11.
ACS Appl Mater Interfaces ; 10(28): 24249-24257, 2018 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-29939714

RESUMEN

Compared to the many studies that focus on the development of novel molecular frameworks pertaining to functionalized fluorescent materials, there is lesser emphasis on side chains even though they have a significant impact on the properties and applications of fluorescent materials. In this study, a series of pyridinium-functionalized tetraphenylethene salts (TPEPy-1 to TPEPy-4) possessing different alkyl chains are synthesized, and the influence of chain length on their optical performance and applications is thoroughly investigated. By changing the alkyl chain, the fluorogens exhibit opposite emission behavior in aqueous media because of their distinct hydrophobic nature, and their solid-state emission can be fine-tuned from green to red owing to their distinct molecular configuration. In addition, by increasing the chain length, the microstructure of the self-assembled fluorogens converts from microplates to microrods with various emission colors. Moreover, TPEPy-1 exhibits dual-mode fluorescence "turn-on" response toward NO3- and ClO4- in aqueous media because the anions induce the self-assembly of fluorogens. Furthermore, the fluorogens display cellular uptake selectivity while the proper alkyl chain impels the fluorogens to penetrate the cell membrane and accumulate in the mitochondria with high specificity.

12.
Endocrinology ; 159(8): 2905-2916, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29850805

RESUMEN

Arterial calcification is a common cardiovascular disease that initiates from a process of osteoblastic differentiation of vascular smooth muscle cells (VSMCs). Accumulating evidence has demonstrated that microRNAs play an important role in regulating arterial calcification. miR-204 was significantly downregulated in calcified human renal arteries from patients with uremia; calcified arteries of mice, due to 5/6 nephrectomy with a high-phosphate diet (5/6 NTP); and in VSMCs induced by high phosphate concentration. The overexpression of miR-204 alleviated the osteoblastic differentiation of VSMCs. Bisulphite sequencing PCR revealed that CpG sites upstream of miR-204 DNA were hypermethylated in calcified VSMCs; in calcified arteries of mice, due to 5/6 NTP; and in calcified renal artery tissues from patients with uremia. Moreover, increased DNMT3a resulted in the hypermethylation of miR-204 in high phosphate concentration-induced VSMCs, whereas 5-aza-2'-deoxycytidine could restore the expression of miR-204 in high phosphate concentration-induced VSMCs. Moreover, we found that DNMT3a was the target of miR-204, and the methylation ratio of miR-204 was decreased significantly, meaning that the expression of miR-204 was restored when DNMT3a was knocked down by using DNMT3a small interfering RNA, resulting in abrogation of the effect of high phosphate concentration on VSMC calcification. The progress of arterial calcification is regulated by the miR-204/DNMT3a regulatory circuit.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , MicroARNs/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Arteria Renal/metabolismo , Calcificación Vascular/metabolismo , Adulto , Animales , Aorta/citología , Estudios de Casos y Controles , Islas de CpG , Metilación de ADN , ADN Metiltransferasa 3A , Regulación hacia Abajo , Femenino , Expresión Génica , Humanos , Técnicas In Vitro , Fallo Renal Crónico/metabolismo , Fallo Renal Crónico/cirugía , Trasplante de Riñón , Donadores Vivos , Masculino , Metilación , Ratones , Persona de Mediana Edad , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Nefrectomía , Osteoblastos , Fosfatos/metabolismo , Arteria Renal/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Uremia/metabolismo
13.
Mol Cell Endocrinol ; 279(1-2): 1-8, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17942216

RESUMEN

WISP3 is essential for maintaining cartilage integrity mainly by regulating the expression of collagen II, and mutations of WISP3 linked to spondyloepiphyseal dysplasia tarda with progressive arthropathy (SEDT-PA) can compromise this function and lead to cartilage loss. The aim of this study was to evaluate the effect of WISP3 on insulin-like growth factor (IGF) signaling in human chondrocytes, investigate whether WISP3 up-regulates collagen II through the IGF signaling pathway, and compare IGF signaling between wild-type and mutant WISP3. Experimental results suggest that WISP3 up-regulates collagen II expression and inhibits the activation of IGF-IR, IRS-1, and ERK kinase in human chondrocytes, and mutation of WISP3 augments IGF signaling in human chondrocytes. In addition to the IGF signaling pathway, WISP3 might up-regulate collagen II expression through an IGF-independent signaling cascade.


Asunto(s)
Condrocitos/metabolismo , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Transducción de Señal/fisiología , Proteínas CCN de Señalización Intercelular , Línea Celular , Condrocitos/efectos de los fármacos , Colágeno/metabolismo , Humanos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/farmacología , Factor I del Crecimiento Similar a la Insulina/efectos de los fármacos , Mutación , Receptor IGF Tipo 1/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
14.
Peptides ; 28(3): 708-18, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17109997

RESUMEN

The aim of this study was to investigate the effects of apelin on proliferation and apoptosis of mouse osteoblastic MC3T3-E1 cells. APJ was expressed in MC3T3-E1 cells. Apelin did not affect Runx2 expression, alkaline phosphatase (ALP) activity, osteocalcin and type I collagen secretion, suggesting that it has no effect on osteoblastic differentiation of MC3T3-E1 cells. However, apelin stimulated MC3T3-E1 cell proliferation and inhibited cell apoptosis induced by serum deprivation. Our study also shows that apelin decreased cytochrome c release and caspase-3, capase-8 and caspase-9 activation in serum-deprived MC3T3-E1 cells. Apelin activated c-Jun N-terminal kinase (JNK) and Akt (phosphatidylinositol 3-kinase downstream effector), and the JNK inhibitor SP600125, the phosphatidylinositol 3-kinase (PI3-K) inhibitor LY294002 or the Akt inhibitor 1L-6-hydroxymethyl-chiro-inositol 2-(R)-2-O-methyl-3-O-octadecylcarbonate (HIMO) inhibited its effects on proliferation and serum deprivation-induced apoptosis. Furthermore, apelin protected against apoptosis induced by the glucocorticoid dexamethasone or TNF-alpha. Apelin stimulates proliferation and suppresses serum deprivation-induced apoptosis of MC3T3-E1 cells and these actions are mediated via JNK and PI3-K/Akt signaling pathways.


Asunto(s)
Proteínas Portadoras/farmacología , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Células 3T3 , Adipoquinas , Animales , Apelina , Receptores de Apelina , Apoptosis/efectos de los fármacos , Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Citocromos c/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Osteoblastos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/efectos de los fármacos
15.
Oncol Lett ; 14(5): 5457-5463, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29098035

RESUMEN

Mammalian sterile 20-like kinase 1 (Mst1) is a major inhibitor of cell proliferation, and is involved in apoptosis, oncogenesis and organ growth via its ubiquitously encoded serine threonine kinase. Previous studies have demonstrated that Mst1 has a tumor suppressor function in human breast cancer. Mst1 deletion or mutation is associated with tumorigenesis, whereas Mst1 overexpression leads to tumor cell apoptosis and decreases proliferation of tumor cells. Our previous study reported the tumor suppressive function of Mst1, and debated Mst1 as a prognostic factor in human breast cancer. In the present study, Mst1 levels were measured in the plasma of patients in order to elucidate their association with overall and disease-free survival. The results of the present study indicated that Mst1 is a strong prognostic and predictive factor in human breast cancer and a promising anticancer target.

16.
Sci Rep ; 7(1): 3549, 2017 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-28615727

RESUMEN

Arterial calcification is a major complication of cardiovascular disease. Oestrogen replacement therapy in postmenopausal women is associated with lower levels of coronary artery calcification, but its mechanism of action remains unclear. Here, we show that oestrogen inhibits the osteoblastic differentiation of vascular smooth muscle cells (VSMCs) in vitro and arterial calcification in vivo by promoting autophagy. Through electron microscopy, GFP-LC3 redistribution, and immunofluorescence analyses as well as measurement of the expression of the autophagosome marker light-chain I/II (LC3I/II) and autophagy protein 5 (Atg5), we show that autophagy is increased in VSMCs by oestrogen in vitro and in vivo. The inhibitory effect of oestrogen on arterial calcification was counteracted by 3-methyladenine (3MA) or knockdown of Atg5 and was increased by rapamycin. Furthermore, the inhibitory effect of oestrogen on arterial calcification and the degree of autophagy induced by oestrogen were blocked by a nonselective oestrogen receptor (ER) antagonist (ICI 182780), a selective oestrogen receptor alpha (ERα) antagonist (MPP), and ERα-specific siRNA. Our data indicate that oestrogen inhibits the osteoblastic differentiation of VSMCs by promoting autophagy through the ERα signalling pathway in vitro and arterial calcification in vivo by increasing autophagy. Our findings provide new insights into the mechanism by which oestrogen contributes to vascular calcification in vitro and in vivo.


Asunto(s)
Arterias/efectos de los fármacos , Arterias/patología , Autofagia , Calcinosis/tratamiento farmacológico , Estrógenos/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Femenino , Humanos , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología
17.
Regul Pept ; 134(2-3): 118-25, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16563531

RESUMEN

OBJECTIVES: Apelin is a recently discovered peptide that is the endogenous ligand for the orphan G-protein-coupled receptor APJ. Adipocytes can express and secrete apelin. The aim of this study was to characterize apelin and APJ expression in human osteoblasts and to investigate the effects of apelin on osteoblasts. RESULTS: Apelin and APJ were expressed in human osteoblasts. Apelin stimulated proliferation of human osteoblasts, but had no effect on alkaline phosphatase (ALP) activity, osteocalcin and type I collagen production in human osteoblasts. Suppression of APJ with small-interfering RNA (siRNA) abolished the apelin-induced cell proliferation. Apelin induced activation of Akt (Phosphatidylinositol-3 kinase downstream effector), but not MAPKs, such as c-jun N-terminal Kinase (JNK), p38 and ERK1/2 in human osteoblasts. This effect was blocked by suppression of APJ with siRNA. Furthermore, LY294002 (PI3 kinase inhibitor) blocked the activation of Akt by apelin and abolished the apelin-induced cell proliferation. CONCLUSIONS: Human osteoblasts express apelin and APJ and apelin enhances human osteoblast proliferation, but has no effect on osteoblast differentiation, and APJ/PI3 kinase/Akt pathway is involved in the proliferation response. These findings suggest that apelin may function as a mitogenic agent for osteoblasts.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Osteoblastos/metabolismo , Receptores Acoplados a Proteínas G/biosíntesis , Adolescente , Adulto , Fosfatasa Alcalina/metabolismo , Apelina , Receptores de Apelina , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cromonas/farmacología , Colágeno Tipo I/metabolismo , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Persona de Mediana Edad , Morfolinas/farmacología , Osteocalcina/metabolismo , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal/efectos de los fármacos
18.
Endocrinology ; 155(2): 558-67, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24248461

RESUMEN

Arterial calcification is a complex and active regulated process, which results from a process of osteoblastic differentiation of vascular smooth muscle cells (VSMCs). Leptin, the product of the ob gene, mainly regulates food intake and energy expenditure and recently has been considered to be correlated with the arterial calcification. However, the mechanisms of the effects of leptin on osteoblastic differentiation of VSMCs are unknown. We used calcifying vascular smooth muscle cells (CVSMCs) as a model to investigate the relationship between leptin and the osteoblastic differentiation of CVSMCs and the signaling pathways involved. Our experiments demonstrated that leptin could increase expression of receptor activator of nuclear factor-κB ligand (RANKL) and bone morphogenetic protein 4 (BMP4), as well as alkaline phosphatase (ALP) activity, runt-related transcription factor 2 expression, calcium deposition, and the formation of mineralized nodules in CVSMCs. Suppression of RANKL with small interfering RNA abolished the leptin-induced ALP activity and BMP4 expression in CVSMCs. Leptin could activate the ERK1/2 and phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. Furthermore, pretreatment with the ERK inhibitor PD98059 and the PI3K inhibitor LY294002 abolished leptin-induced RANKL expression and blocked the promotion of ALP activity of CVSMCs. Silencing of the leptin receptor OB-Rb with small interfering RNA abolished leptin-induced activation of ERK and Akt and the expression of RANKL and reversed the effects of leptin on ALP activity. Meanwhile, addition of Noggin (the BMP4 inhibitor) blunted the effect of leptin on ALP activity. These results show that leptin can promote osteoblastic differentiation of CVSMCs by the OB-Rb/ERK1/2/RANKL-BMP4 and OB-Rb/PI3K/Akt/RANKL-BMP4 pathways.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Leptina/farmacología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Osteoblastos/citología , Ligando RANK/metabolismo , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Calcinosis/metabolismo , Calcio/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Femenino , Ratones , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Ligando RANK/genética , Receptores de Leptina/genética , Receptores de Leptina/metabolismo
19.
J Photochem Photobiol B ; 118: 58-65, 2013 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-23218540

RESUMEN

Photochemical properties and phototoxicity of Pazufloxacin (PAX) were systematically investigated in aqueous solutions using UV-Vis, fluorescence, laser flash photolysis, pulse radiolysis and SDS-PAGE gel electrophoresis techniques. PAX triplet-state ((3)PAX(*)) absorption spectra (λ(max)=570 nm) was determined. (3)PAX(*) was quenched by PAX and O(2), with rate constants of 6.9×10(8) and 3.2×10(8) dm(3) mol(-1) s(-1), respectively. The pK(a) values (5.7 and 8.6) for the protonation equilibrium were determined by UV-Vis and fluorescence techniques. The PAX triplet energy (E(T)=260.3 kJ/mol) was obtained using energy transfer method. The reaction of electron transfer from tryptophan (TrpH) and dGMP to (3)PAX(*) was found with rate constants of 8.8×10(7) and 8.7×10(6) dm(3) mol(-1) s(-1), respectively. The rate constants for reactions of ()OH, SO(4)(-) and hydrated electron with PAX were found to be 5.8×10(8), 2.1×10(9) and 9×10(9)d m(3) mol(-1) s(-1), respectively. Based on the results obtained, a rational scheme for dGMP, TrpH and lysozyme photodamage induced by PAX was proposed.


Asunto(s)
Fluoroquinolonas/química , Fluoroquinolonas/efectos de la radiación , Oxazinas/química , Oxazinas/efectos de la radiación , Nucleótidos de Desoxiguanina/química , Transporte de Electrón , Electroforesis en Gel de Poliacrilamida , Fluoroquinolonas/toxicidad , Rayos Láser , Muramidasa/efectos de los fármacos , Oxazinas/toxicidad , Procesos Fotoquímicos , Fotólisis , Radiólisis de Impulso , Triptófano/efectos de los fármacos
20.
Int J Endocrinol ; 2013: 368970, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23606838

RESUMEN

It has been presumed that adipokines deriving from adipose tissue may play important roles in bone metabolism. Omentin-1, a novel adipokine, which is selectively expressed in visceral adipose tissue, has been reported to stimulate proliferation and inhibit differentiation of mouse osteoblast. However, little information refers to the effect of omentin-1 on human osteoblast (hOB) proliferation. The current study examined the potential effects of omentin-1 on proliferation in hOB and the signal pathway involved. Omentin-1 promoted hOB proliferation in a dose-dependent manner as determined by [(3)H]thymidine incorporation. Western blot analysis revealed that omentin-1 induced activation of Akt (phosphatidylinositol-3 kinase downstream effector) and such effect was impeded by transfection of hOB with Akt-siRNA. Furthermore, LY294002 (a selective PI3K inhibitor) and HIMO (a selective Akt inhibitor) abolished the omentin-1-induced hOB proliferation. These findings indicate that omentin-1 induces hOB proliferation via the PI3K/Akt signaling pathway and suggest that osteoblast is a direct target of omentin-1.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA