Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Cell Mol Med ; 25(12): 5486-5496, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33955654

RESUMEN

This study aimed to investigate the molecular mechanisms underlying the role of bone marrow mesenchymal stem cells (BMMSCs)-derived exosomes in ischaemia/reperfusion (IR)-induced damage, and the role of oridonin in the treatment of IR. Exosomes were isolated from BMMSCs. Western blot analysis was done to examine the expression of proteins including CD63, CD8, apoptotic-linked gene product 2 interacting protein X (AliX), Beclin-1, ATG13, B-cell lymphoma-2 (Bcl-2), apoptotic peptidase activating factor 1 (Apaf1) and Bcl2-associated X (Bax) in different treatment groups. Accordingly, the expression of CD63, CD81 and AliX was higher in BMMSCs-EXOs and IR + BMMSCs-EXOs + ORI groups compared with that in the BMMSCs group. And BMMSCs-derived exosomes inhibited the progression of IR-induced myocardial damage, while this protective effect was boosted by the pre-treatment with oridonin. Moreover, Beclin-1, ATG13 and Bcl-2 were significantly down-regulated while Apaf1 and Bax were significantly up-regulated in IR rats. And the presence of BMMSCs-derived exosomes partly alleviated IR-induced dysregulation of these proteins, while the oridonin pre-treatment boosted the effect of these BMMSCs-derived exosomes. The inhibited proliferation and promoted apoptosis of H9c2 cells induced by hypoxia/reperfusion (HR) were mitigated by the administration of BMMSCs-derived exosomes. Meanwhile, HR also induced down-regulation of Beclin-1, ATG13 and Bcl-2 expression and up-regulation of Apaf1 and Bax, which were mitigated by the administration of BMMSCs-derived exosomes. And oridonin pre-treatment boosted the effect of BMMSCs-derived exosomes. In conclusion, our results validated that BMMSCs-derived exosomes suppressed the IR-induced damages by participating in the autophagy process, while the pre-treatment with oridonin could boost the protective effect of BMMSCs-derived exosomes.


Asunto(s)
Apoptosis , Autofagia , Diterpenos de Tipo Kaurano/farmacología , Exosomas/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Daño por Reperfusión Miocárdica/terapia , Animales , Exosomas/efectos de los fármacos , Masculino , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Ratas , Ratas Sprague-Dawley
2.
J Cell Mol Med ; 24(17): 9890-9897, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32790060

RESUMEN

Methionine restrictive diet may alleviate ischaemia/reperfusion (I/R)-induced myocardial injury, but its underlying mechanism remains unclear. HE staining was performed to evaluate the myocardial injury caused by I/R and the effect of methionine-restricted diet (MRD) in I/R mice. IHC and Western blot were carried out to analyse the expression of CSE, CHOP and active caspase3 in I/R mice and hypoxia/reoxygenation (H/R) cells. TUNEL assay and flow cytometry were used to assess the apoptotic status of I/R mice and H/R cells. MTT was performed to analyse the proliferation of H/R cells. H2S assay was used to evaluate the concentration of H2S in the myocardial tissues and peripheral blood of I/R mice. I/R-induced mediated myocardial injury and apoptosis were partially reversed by methionine-restricted diet (MRD) via the down-regulation of CSE expression and up-regulation of CHOP and active caspase3 expression. The decreased H2S concentration in myocardial tissues and peripheral blood of I/R mice was increased by MRD. Accordingly, in a cellular model of I/R injury established with H9C2 cells, cell proliferation was inhibited, cell apoptosis was increased, and the expressions of CSE, CHOP and active caspase3 were dysregulated, whereas NaHS treatment alleviated the effect of I/R injury in H9C2 cells in a dose-dependent manner. This study provided a deep insight into the mechanism underlying the role of MRD in I/R-induced myocardial injury.


Asunto(s)
Lesión Renal Aguda/metabolismo , Diabetes Mellitus Experimental/dietoterapia , Metionina/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Lesión Renal Aguda/complicaciones , Lesión Renal Aguda/dietoterapia , Animales , Apoptosis/genética , Caspasa 3/genética , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Humanos , Riñón , Metionina/genética , Ratones , Ratones Endogámicos NOD/genética , Ratones Endogámicos NOD/metabolismo , MicroARNs/genética , Daño por Reperfusión Miocárdica/dietoterapia , Daño por Reperfusión Miocárdica/etiología , Miocardio/metabolismo , Daño por Reperfusión/complicaciones , Daño por Reperfusión/dietoterapia , Daño por Reperfusión/metabolismo , Transducción de Señal/genética , Sulfitos/farmacología , Factor de Transcripción CHOP/genética
3.
Med Sci Monit ; 24: 1276-1281, 2018 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-29499032

RESUMEN

BACKGROUND The incidence rate of thyroid cancer has increased greatly during the last few decades, and highly sensitive and specific methods for early diagnosis and prognostic evaluation remain lacking. In this study, we investigated a novel approach based on microwave theory to detect thyroid cancer. MATERIAL AND METHODS Freshly excised thyroid tissues (n=236) from 48 patients were identified as normal or malignant using histology. Each sample was measured for effective dielectric permittivity and effective conductivity (0.5-8 GHz). The means of each of these parameters of the normal and malignant groups were compared. RESULTS The effective dielectric permittivities of normal and malignant thyroid tissues were 24.026±1.951 to 17.950±1.648 and 69.782±2.734 to 57.356±1.802, respectively. Also, as a function of frequency, the effective conductivities of normal and malignant thyroid cancer were 0.8395±0.2013 to 1.8730±0.0979 and 1.8960±0.5024 to 9.7461±0.9349 (S/m), respectively. The mean effective dielectric permittivities and effective conductivities of normal thyroid tissues were significantly lower than that of thyroid cancer tissues. CONCLUSIONS Measuring the effective dielectric permittivity and effective conductivity of excised thyroid tissues may be a new and viable method to determine malignancy in thyroid cancer.


Asunto(s)
Espectroscopía Dieléctrica/métodos , Glándula Tiroides/citología , Neoplasias de la Tiroides/diagnóstico , China , Conductividad Eléctrica , Impedancia Eléctrica , Femenino , Humanos , Masculino , Microondas/uso terapéutico , Neoplasias/diagnóstico
4.
Biomacromolecules ; 18(9): 2820-2829, 2017 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-28731675

RESUMEN

Following myocardial infarction (MI), degradation of extracellular matrix (ECM) by upregulated matrix metalloproteinases (MMPs) especially MMP-2 decreases tissue mechanical properties, leading to cardiac function deterioration. Attenuation of cardiac ECM degradation at the early stage of MI has the potential to preserve tissue mechanical properties, resulting in cardiac function increase. Yet the strategy for efficiently preventing cardiac ECM degradation remains to be established. Current preclinical approaches have shown limited efficacy because of low drug dosage allocated to the heart tissue, dose-limiting side effects, and cardiac fibrosis. To address these limitations, we have developed a MMP-2 inhibitor delivery system that can be specifically delivered into infarcted hearts at early stage of MI to efficiently prevent MMP-2-mediated ECM degradation. The system was based on an injectable, degradable, fast gelation, and thermosensitive hydrogel, and a MMP-2 specific inhibitor, peptide CTTHWGFTLC (CTT). The use of fast gelation hydrogel allowed to completely retain CTT in the heart tissue. The system was able to release low molecular weight CTT over 4 weeks possibly due to the strong hydrogen bonding between the hydrogel and CTT. The release kinetics was modulated by amount of CTT loaded into the hydrogel, and using chondroitin sulfate and heparin that can interact with CTT and the hydrogel. Both glycosaminoglycans augmented CTT release, while heparin more greatly accelerated the release. After it was injected into the infarcted hearts for 4 weeks, the released CTT efficiently prevented cardiac ECM degradation as it not only increased tissue thickness but also preserved collagen composition similar to that in the normal heart tissue. In addition, the delivery system significantly improved cardiac function. Importantly, the delivery system did not induce cardiac fibrosis. These results demonstrate that the developed MMP-2 inhibitor delivery system has potential to efficiently reduce adverse myocardial remodeling and improve cardiac function.


Asunto(s)
Portadores de Fármacos/síntesis química , Liberación de Fármacos , Hidrogeles/síntesis química , Inhibidores de la Metaloproteinasa de la Matriz/farmacocinética , Infarto del Miocardio/tratamiento farmacológico , Péptidos Cíclicos/farmacocinética , Animales , Sulfatos de Condroitina/química , Portadores de Fármacos/efectos adversos , Portadores de Fármacos/química , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Heparina/química , Humanos , Hidrogeles/efectos adversos , Hidrogeles/química , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/administración & dosificación , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Péptidos Cíclicos/administración & dosificación , Péptidos Cíclicos/uso terapéutico , Ratas , Ratas Sprague-Dawley
5.
J Cell Mol Med ; 20(1): 29-37, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26282458

RESUMEN

Mesenchymal stem cell-based therapy has emerged as a promising approach for the treatment of peripheral arterial disease. The purpose of this study was to examine the potential effects of human placenta-derived mesenchymal stem cells (PMSCs) on mouse hindlimb ischemia. PMSCs were isolated from human placenta tissue and characterized by flow cytometry. An in vivo surgical ligation-induced murine limb ischemia model was generated with fluorescent dye (CM-DiI) labelled PMSCs delivered via intramuscular injection. Our data show that PMSCs treatment significantly enhanced microvessel density, improved blood perfusion and diminished pathologies in ischemic mouse hindlimbs as compared to those in the control group. Further immunostaining studies suggested that injected PMSCs can incorporate into the vasculature and differentiate into endothelial and smooth muscle cells to enhance angiogenesis in ischemic hind limbs. This may in part explain the beneficial effects of PMSCs treatment. Taken together, we found that PMSCs treatment might be an effective treatment modality for treatment of ischemia-induced injury to mouse hind limbs by enhancement of angiogenesis.


Asunto(s)
Miembro Posterior/irrigación sanguínea , Isquemia/terapia , Trasplante de Células Madre Mesenquimatosas , Neovascularización Fisiológica , Animales , Diferenciación Celular , Células Cultivadas , Citocinas/metabolismo , Células Endoteliales/metabolismo , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Placenta/citología , Embarazo
6.
J Biol Chem ; 290(40): 24592-603, 2015 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-26306047

RESUMEN

Cell membrane repair is an important aspect of physiology, and disruption of this process can result in pathophysiology in a number of different tissues, including wound healing, chronic ulcer and scarring. We have previously identified a novel tripartite motif family protein, MG53, as an essential component of the cell membrane repair machinery. Here we report the functional role of MG53 in the modulation of wound healing and scarring. Although MG53 is absent from keratinocytes and fibroblasts, remarkable defects in skin architecture and collagen overproduction are observed in mg53(-/-) mice, and these animals display delayed wound healing and abnormal scarring. Recombinant human MG53 (rhMG53) protein, encapsulated in a hydrogel formulation, facilitates wound healing and prevents scarring in rodent models of dermal injuries. An in vitro study shows that rhMG53 protects against acute injury to keratinocytes and facilitates the migration of fibroblasts in response to scratch wounding. During fibrotic remodeling, rhMG53 interferes with TGF-ß-dependent activation of myofibroblast differentiation. The resulting down-regulation of α smooth muscle actin and extracellular matrix proteins contributes to reduced scarring. Overall, these studies establish a trifunctional role for MG53 as a facilitator of rapid injury repair, a mediator of cell migration, and a modulator of myofibroblast differentiation during wound healing. Targeting the functional interaction between MG53 and TGF-ß signaling may present a potentially effective means for promoting scarless wound healing.


Asunto(s)
Proteínas Portadoras/fisiología , Membrana Celular/metabolismo , Proteínas Musculares/fisiología , Proteínas de Transporte Vesicular/fisiología , Cicatrización de Heridas/fisiología , Células 3T3 , Actinas/metabolismo , Animales , Diferenciación Celular , Movimiento Celular , Cicatriz/patología , Colágeno Tipo I/metabolismo , Fibroblastos/citología , Fibronectinas/metabolismo , Fibrosis/patología , Regulación de la Expresión Génica , Humanos , Hidrogeles/química , Queratinocitos/metabolismo , Proteínas de la Membrana , Ratones , Músculo Liso/metabolismo , Miofibroblastos/metabolismo , Conejos , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Piel/patología , Proteínas de Motivos Tripartitos
7.
Med Sci Monit ; 22: 1161-70, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27054781

RESUMEN

BACKGROUND Denervation-induced skeletal muscle atrophy results in significant biochemical and physiological changes potentially leading to devastating outcomes including increased mortality. Effective treatments for skeletal muscle diseases are currently not available. Muscle-specific miRNAs, such as miR-206, play an important role in the regulation of muscle regeneration. The aim of the present study was to examine the beneficial effects of miR-206 treatment during the early changes in skeletal muscle atrophy, and to study the underlying signaling pathways in a rat skeletal muscle atrophy model. MATERIAL AND METHODS The rat denervation-induced skeletal muscle atrophy model was established. miRNA-206 was overexpressed with or without TGF-ß1 inhibitor in the rats. The mRNA and protein expression of HDAC4, TGF-ß1, and Smad3 was determined by real-time PCR and western blot. The gastrocnemius muscle cross-sectional area and relative muscle mass were measured. MyoD1, TGF-ß1, and Pax7 were determined by immunohistochemical staining. RESULTS After sciatic nerve surgical transection, basic muscle characteristics, such as relative muscle weight, deteriorated continuously during a 2-week period. Injection of miR-206 (30 µg/rat) attenuated morphological and physiological deterioration of muscle characteristics, prevented fibrosis effectively, and inhibited the expression of TGF-ß1 and HDAC4 as assessed 2 weeks after denervation. Moreover, miR-206 treatment increased the number of differentiating (MyoD1+/Pax7+) satellite cells, thereby protecting denervated muscles from atrophy. Interestingly, the ability of miR-206 to govern HDAC4 expression and to attenuate muscle atrophy was weakened after pharmacological blockage of the TGF-b1/Smad3 axis. CONCLUSIONS TGF-ß1/Smad3 signaling pathway is one of the crucial signaling pathways by which miR-206 counteracts skeletal muscle atrophy by affecting proliferation and differentiation of satellite cells. miR-206 may be a potential target for development of a new strategy for treatment of patients with early denervation-induced skeletal muscle atrophy.


Asunto(s)
Histona Desacetilasas/metabolismo , MicroARNs/administración & dosificación , Músculo Esquelético/inervación , Músculo Esquelético/patología , Atrofia Muscular/terapia , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Desnervación , Histona Desacetilasas/genética , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/etiología , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Mioblastos/metabolismo , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Satélite del Músculo Esquelético/metabolismo , Nervio Ciático/cirugía , Transducción de Señal , Proteína smad3/genética , Factor de Crecimiento Transformador beta1/genética
8.
Am J Physiol Heart Circ Physiol ; 308(1): H11-7, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25380816

RESUMEN

Palmitate (PA) impairs endothelial progenitor cells (EPCs). However, the molecular mechanism underlying the suppressive function of PA remains largely unknown. Ceramide, a free fatty acid metabolite, mediates multiple cellular signals. We hypothesized that ceramide acts as an intermediate molecule to mediate inhibition of EPCs by PA. We first demonstrated that PA could inhibit the attachment, migration, and tube formation of EPCs through suppression of the Akt/endothelial nitric oxide (NO) synthase (eNOS) signaling pathway. In addition, we observed that PA could induce ceramide accumulation in EPCs. To test whether the accumulation of ceramide causes EPC dysfunction, the ceramide synthesis inhibitors myriocin and fumonisin B1 were used. We that found both inhibitors could effectively abolish PA-mediated EPC inhibition. Furthermore, the ceramide deacylation inhibitor N-oleoylethanolamine could augment the inhibitory effect of PA on EPCs, indicating that it is ceramide, not its metabolites, that mediates the suppression of EPCs by PA. We have previously shown that Akt/eNOS phosphorylation was reduced after PA treatment, which, in turn, hampered the normal bioavailability of NO, leading to impaired functions of EPCs. To test the role for ceramide in this process, a clinically used NO donor, sodium nitroprusside, was used. We found that sodium nitroprusside could rescue the suppressive effects of ceramide on EPCs, suggesting that ceramide-mediated EPC inhibition might be through reduction of NO production. Taken together, our findings indicated that ceramide-induced reduction of NO might be the molecular mechanism for PA-mediated EPC inhibition; thus, targeting either ceramide or NO production might be an effective means for improvement of EPC functions in diseases.


Asunto(s)
Ceramidas/metabolismo , Células Progenitoras Endoteliales/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ácido Palmítico/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Células Progenitoras Endoteliales/enzimología , Inhibidores Enzimáticos/farmacología , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Fosforilación , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
9.
Cell Physiol Biochem ; 35(1): 353-63, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25591776

RESUMEN

BACKGROUND/AIMS: Bone marrow (BM)-derived endothelial progenitor cells (EPCs) play a critical role in angiogenesis and vascular repair. Some environmental insults, like fine particulate matter (PM) exposure, significantly impair cardiovascular functions. However, the mechanisms for PM-induced adverse effects on cardiovascular system remain largely unknown. The present research was to study the detrimental effects of PM on EPCs and explore the potential mechanisms. METHODS: PM was intranasal-distilled into male C57BL/6 mice for one month. Flow cytometry was used to measure the number of EPCs, apoptosis level of circulating EPCs and intracellular reactive oxygen species (ROS) formation. Serum TNF-α and IL-1ß were measured using ELISA. To determine the role of PM-induced ROS in EPC apoptosis, PM was co-administrated with the antioxidant N-acetylcysteine (NAC) in wild type mice or used in a triple transgenic mouse line (TG) with overexpression of antioxidant enzyme network (AON) composed of superoxide dismutase (SOD)1, SOD3, and glutathione peroxidase (Gpx-1) with decreased in vivo ROS production. RESULTS: PM treatment significantly decreased circulating EPC population, promoted apoptosis of EPCs in association with increased ROS production and serum TNF-α and IL-1ß levels, which could be effectively reversed by either NAC treatment or overexpression of AON. CONCLUSION: PM exposure significantly decreased circulating EPCs population due to increased apoptosis via ROS formation in mice.


Asunto(s)
Apoptosis/efectos de los fármacos , Material Particulado/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Acetilcisteína/farmacología , Animales , Células de la Médula Ósea/citología , Células Cultivadas , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Ensayo de Inmunoadsorción Enzimática , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Interleucina-1beta/sangre , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Factor de Necrosis Tumoral alfa/sangre
10.
Cell Physiol Biochem ; 34(6): 1998-2006, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25562148

RESUMEN

BACKGROUND: Peripheral artery disease (PAD) is a major health burden in the world. Stem cell-based therapy has emerged as an attractive treatment option in regenerative medicine. In this study, we sought to test the hypothesis that stem cell-based therapy can ameliorate ischemia induced limb injury. METHODS: We isolated mesenchymal stem cells derived from human placentas (PMSCs) and intramuscularly transplanted them into injured hind limbs. Treatment with PMSCs reduced acute muscle fibers apoptosis induced by ischemia. RESULTS: PMSC treatment significantly enhanced regeneration of the injured hind limb by reducing fibrosis and enhancing running capacity when the animals were subjected to treadmill training. Mechanistically, injected PMSCs can modulate acute inflammatory responses by reducing neutrophil and macrophage infiltration following limb ischemia. ELISA assays further confirmed that PMSC treatment can also reduce pro-inflammatory cytokines, TNF-α and IL-6, and enhance anti-inflammatory cytokine, IL-10 at the injury sites. CONCLUSION: Taken together, our results demonstrated that PMSCs can be a potential effective therapy for treatment of PAD via immunomodulation.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Trasplante de Células Madre Mesenquimatosas , Enfermedad Arterial Periférica/terapia , Daño por Reperfusión/terapia , Animales , Extremidades/lesiones , Femenino , Humanos , Inmunomodulación , Ratones , Enfermedad Arterial Periférica/inmunología , Enfermedad Arterial Periférica/patología , Placenta/citología , Placenta/inmunología , Embarazo , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología
11.
Biochem Biophys Res Commun ; 452(1): 191-6, 2014 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-25019980

RESUMEN

The stromal cell-derived factor-1α/C-X-C chemokine receptor 4 (SDF-1/CXCR4) axis is involved in various aspects of tissue repair, regeneration and development. However, the role of SDF-1/CXCR4 in acute lung injury (ALI) remains largely unknown. The aim of the present investigation is to examine pathological changes in a rabbit model with ALI induced by oleic acid (OA) and to explore the protective effect of SDF-1α on ALI. Intravenous application (i.v.) of oleic acid (0.1 ml/kg/h for 2h) provoked pulmonary hemorrhage, edema, and protein leakage, resulting in severe ALI. When the rabbit received an infusion of SDF-1α (20 µg/kg/24h) for 30 min before OA treatment, SDF-1α seemed to significantly improve the pathologies associated with OA-induced ALI. While dissecting the molecular mechanisms underlying the beneficial effects of SDF-1α, we found that SDF-1/CXCR4 is expressed in uninjured lung tissues but is greatly reduced after OA treatment. Interestingly, intravenous delivery of SDF-1α could target an injured lung and rescue expression of CXCR4, which in turn activates anti-apoptotic proteins, Bcl-1 and Bcl-xl, but does not affect pro-apoptotic proteins, such as Bad and Bax. These data suggested that SDF-1α could protect rabbit lungs from AIL. The molecular mechanism might be associated with upregulating anti-apoptosis family expression through CXCR4. Thus, SDF-1/CXCR4 signaling pathway may be a promising target for treatment of patients with ALI.


Asunto(s)
Quimiocina CXCL12/fisiología , Pulmón/efectos de los fármacos , Ácido Oléico/toxicidad , Animales , Conejos
12.
J Cardiovasc Pharmacol ; 62(2): 143-53, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23609327

RESUMEN

AIM: The aim of this study was to explore the mechanisms underlying the effects of globular adiponectin (gAd) on myocardial ischemia/reperfusion (I/R) injury. METHODS: An in vivo myocardial I/R model and an in vitro neonatal rat cardiomyocyte hypoxia/reoxygenation (H/R) model simulating I/R injury in vivo were adopted to investigate whether and how the cardioprotective effects of gAd are mediated by the inhibition of endoplasmic reticulum (ER) stress. RESULTS: gAd (1 µg/g, intravenously) attenuated the myocardial infarct size, myocardial enzyme activity, and apoptosis in rats with I/R, and similar protection was observed in primary cultures of neonatal rat cardiomyocytes. The protective effects of gAd were associated with the suppression of ER stress, as evidenced by reversing the upregulation of 78-kDa glucose-regulated protein, C/EBP homologous protein, and caspase-12 that were induced by H/R and thapsigargin. In addition, gAd conferred resistance to ER stress and cardiomyocyte injury by modulating ER Ca²âº-ATPase (SERCA) activity. Moreover, gAd further increased H/R-enhanced Akt phosphorylation. The protective effects of gAd on ER stress and SERCA activity were abolished by preincubation of rat neonatal cardiomyocytes with the PI3K inhibitor LY294002. Consistent with this finding, I/R-induced ER stress and SERCA dysfunction were also significantly ameliorated by gAd. These effects involved PI3K/Akt signaling pathway. CONCLUSIONS: The protective effects of gAd during I/R are mediated, at least in part, by modulating SERCA activity and consequently suppressing ER stress via the activation of PI3K/Akt signaling.


Asunto(s)
Adiponectina/uso terapéutico , Cardiotónicos/uso terapéutico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Isquemia Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/prevención & control , Fragmentos de Péptidos/uso terapéutico , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Adiponectina/administración & dosificación , Adiponectina/genética , Adiponectina/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Cardiotónicos/administración & dosificación , Cardiotónicos/farmacología , Hipoxia de la Célula , Células Cultivadas , Corazón/efectos de los fármacos , Infusiones Intravenosas , Masculino , Infarto del Miocardio/prevención & control , Isquemia Miocárdica/enzimología , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Miocardio/citología , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/química , Transducción de Señal/efectos de los fármacos
13.
Aging (Albany NY) ; 13(8): 11942-11953, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33875621

RESUMEN

Atherosclerosis is a chronic inflammatory disease known to be mediated by numerous factors, among which endothelial dysfunction plays a critical role. Oscillatory shear stress induces endothelial cells to lose their anti-atherosclerotic properties and downregulates the expression of the innate protective transcription factor, Krüppel-like factor 2 (KLF2), which is typically upregulated in vascular endothelial cells in response to harmful stimuli. Oxidative stress and inflammation impair endothelial function and damage their survival. Oscillatory shear stress also promotes generation of reactive oxygen species and production of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), thereby further promoting endothelial dysfunction and formation of atherosclerotic plaque. A major event in the development of atherosclerotic plaque is rolling and adhesion of monocytes to endothelial cells, which is mediated by adhesion molecules including vascular cellular adhesion molecule 1 and endothelial-selectin. Expression of these molecules is also upregulated by oscillatory shear stress. Estrogen has long been recognized as a protective agent against atherosclerosis, but the mechanisms through which estrogen receptors prevent atherogenesis remain unclear. In the present study, we investigated the role of the G-coupled protein estrogen receptor (GPR30) in oscillatory shear stress- induced endothelial dysfunction. We show that agonism of GPR30 by its specific agonist G1 prevented oscillatory shear stress -induced oxidative stress markers and production of inflammatory cytokines and adhesion molecules. As a result, GPR30 activation suppresses monocytes adhesion to endothelial cells. Furthermore, we demonstrate that GPR30 prevents oscillatory shear stress- induced downregulation of KLF2 via ERK5 pathway. These findings suggest that endothelial GPR30 is potential target to suppress oscillatory shear stress mediated atherogenesis.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Adhesión Celular/efectos de los fármacos , Factores de Transcripción de Tipo Kruppel/metabolismo , Quinolinas/farmacología , Receptores Acoplados a Proteínas G/agonistas , Aterosclerosis/patología , Adhesión Celular/genética , Selectina E/metabolismo , Células Endoteliales , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Quinolinas/uso terapéutico , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Estrés Mecánico , Células THP-1 , Molécula 1 de Adhesión Celular Vascular/metabolismo
14.
Front Cardiovasc Med ; 8: 638663, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33796571

RESUMEN

Background: Low-density lipoprotein cholesterol (LDL-C) is a critical surrogate outcome for cardiovascular disease (CVD). Recent observational studies identified different predictive lipid parameters, but these have not been fully validated in the Chinese population. This study aimed to compare the predictive value of lipid parameters for cardiovascular outcomes in Chinese statin-naïve patients who underwent percutaneous coronary intervention (PCI). Methods: We retrospectively recruited statin-naïve patients who underwent PCI for stable angina and acute coronary syndrome at Sichuan Provincial People's Hospital between 1 January 2016 and 31 December 2017. A follow-up was conducted via outpatient visits or telephone. We divided patients into three groups based on lipid parameter tertiles. We calculated the hazard ratios (HRs) of the highest and lowest tertiles for major adverse cardiovascular events (MACEs) using multivariate Cox proportional hazards regression. We compared the association strength of lipid parameters with MACEs using the HR of non-LDL-C lipid parameters relative to LDL-C. Results: Among 445 included patients, the highest LDL-C, LDL-C/high-density lipoprotein cholesterol (HDL-C), atherosclerosis index, and non-HDL-C level tertiles were associated with an average increase of 165% (HR 2.65, confidence interval [CI] 1.26 to 5.61; P = 0.01), 324% (HR 4.24, CI 1.89 to 9.52; P < 0.001), 152% (HR 2.52, CI 1.22 to 5.22; P = 0.01), and 125% (HR 2.25, CI 1.09 to 4.64; P = 0.01) in the hazard of composite CVD, respectively. Lipoprotein (a) levels did not show a significant association with the endpoints. Except for LDL-C/HDL-C, different lipid parameter HR ratios were <1.0; none were statistically significant. Conclusion: Compared with non-LDL-C lipid parameters, LDL-C acts better predictive value for cardiovascular outcomes in general Chinese statin-naïve post-PCI patients.

15.
Thorac Cancer ; 9(4): 459-465, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29465782

RESUMEN

BACKGROUND: Despite the high incidence of breast cancer worldwide, methods for early non-invasive diagnosis and sensitive and specific prognostic evaluation remain difficult. In this study, we investigated microwave parameters as a potential non-invasive approach to detect breast cancer. METHODS: Samples of freshly excised breast tissues (n = 509) from 98 patients were identified as normal, benign tumor, or malignant cancer via histology. Further samples were prepared and the microwave effective dielectric permittivity and effective conductivity were measured every 0.0375 GHz from 0.5 GHz to 8 GHz. These parameters were compared among the breast tissue types. RESULTS: The effective relative permittivity and effective conductivity at each frequency was significantly higher in breast cancer tissues compared with benign tumors, which in turn was significantly higher than in normal breast tissue. The standard deviation of each parameter was narrowest at ~2.5 GHz in both normal and malignant breast tissues. CONCLUSIONS: The effective dielectric permittivity and effective conductivity, measured via microwave technology, could differentiate breast cancer from normal and benign tumor tissues.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Detección Precoz del Cáncer , Invasividad Neoplásica/diagnóstico , Neoplasias/diagnóstico , Neoplasias de la Mama/fisiopatología , Conductividad Eléctrica , Femenino , Humanos , Microondas , Invasividad Neoplásica/fisiopatología , Neoplasias/fisiopatología , Pronóstico
17.
Acta Biomater ; 31: 99-113, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26689466

RESUMEN

Stem cell therapy is one of the most promising strategies to restore blood perfusion and promote muscle regeneration in ischemic limbs. Yet its therapeutic efficacy remains low owing to the inferior cell survival under the low oxygen and nutrient environment of the injured limbs. To increase therapeutic efficacy, high rates of both short- and long-term cell survival are essential, which current approaches do not support. In this work, we hypothesized that a high rate of short-term cell survival can be achieved by introducing a prosurvival environment into the stem cell delivery system to enhance cell survival before vascularization is established; and that a high rate of long-term cell survival can be attained by building a proangiogenic environment in the system to quickly vascularize the limbs. The system was based on a biodegradable and thermosensitive poly(N-Isopropylacrylamide)-based hydrogel, a prosurvival and proangiogenic growth factor bFGF, and bone marrow-derived mesenchymal stem cells (MSCs). bFGF can be continuously released from the system for 4weeks. The released bFGF significantly improved MSC survival and paracrine effects under low nutrient and oxygen conditions (0% FBS and 1% O2) in vitro. The prosurvival effect of the bFGF on MSCs was resulted from activating cell Kruppel-like factor 4 (KLF4) pathway. When transplanted into the ischemic limbs, the system dramatically improved MSC survival. Some of the engrafted cells were differentiated into skeletal muscle and endothelial cells, respectively. The system also promoted the proliferation of host cells. After only 2weeks of implantation, tissue blood perfusion was completely recovered; and after 4weeks, the muscle fiber diameter was restored similarly to that of the normal limbs. These pronounced results demonstrate that the developed stem cell delivery system has a potential for ischemic limb regeneration. STATEMENT OF SIGNIFICANCE: Stem cell therapy is a promising strategy to restore blood perfusion and promote muscle regeneration in ischemic limbs. Yet its therapeutic efficacy remains low owing to the inferior cell survival under the ischemic environment of the injured limbs. To increase therapeutic efficacy, high rate of cell survival is essential, which current approaches do not support. In this work, we tested the hypothesis that a stem cell delivery system that can continuously release a prosurvival and proangiogenic growth factor will promote high rates of cell survival in the ischemic limbs. The prosurvival effect could augment cell survival before vascularization is established, while the proangiogenic effect could stimulate quick angiogenesis to achieve long-term cell survival. Meanwhile, the differentiation of stem cells into endothelial and myogenic lineages, and cell paracrine effects will enhance vascularization and muscle regeneration.


Asunto(s)
Extremidades/patología , Isquemia/patología , Neovascularización Fisiológica , Regeneración , Células Madre/citología , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Hidrogeles/química , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Oxígeno/química , Perfusión
18.
PLoS One ; 10(6): e0127309, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26058063

RESUMEN

AIMS: Some environmental insults, such as fine particulate matter (PM) exposure, significantly impair the function of stem cells. However, it is unknown if PM exposure could affect the population of bone marrow stem cells (BMSCs). The present study was to investigate the effects of PM on BMSCs population and related mechanism(s). MAIN METHEODS: PM was intranasally distilled into male C57BL/6 mice for one month. Flow cytometry with antibodies for BMSCs, Annexin V and BrdU ware used to determine the number of BMSCs and the levels of their apoptosis and proliferation in vivo. Phosphorylated Akt (P-Akt) level was determined in the BM cells with western blotting. Intracellular reactive oxygen species (ROS) formation was quantified using flow cytometry analysis. To determine the role of PM-induced ROS in BMSCs population, proliferation, and apotosis, experiments were repeated using N-acetylcysteine (NAC)-treated wild type mice or a triple transgenic mouse line with overexpression of antioxidant network (AON) composed of superoxide dismutase (SOD)1, SOD3, and glutathione peroxidase-1 with decreased in vivo ROS production. KEY FINDINGS: PM treatment significantly reduced BMSCs population in association with increased ROS formation, decreased P-Akt level, and inhibition of proliferation of BMSCs without induction of apoptosis. NAC treatment or AON overexpression with reduced ROS formation effectively prevented PM-induced reduction of BMSCs population and proliferation with partial recovery of P-Akt level. SIGNIFICANCE: PM exposure significantly decreased the population of BMSCs due to diminished proliferation via ROS-mediated mechanism (could be partially via inhibition of Akt signaling).


Asunto(s)
Células de la Médula Ósea/citología , Material Particulado/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Células Madre/metabolismo , Acetilcisteína/farmacología , Animales , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Espacio Intracelular/metabolismo , Masculino , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA