Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Inorg Chem ; 63(15): 6787-6797, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38556762

RESUMEN

The electrocatalytic reduction of NO2- (NO2RR) holds promise as a sustainable pathway to both promoting the development of emerging NH3 economies and allowing the closing of the NOx loop. Highly efficient electrocatalysts that could facilitate this complex six-electron transfer process are urgently desired. Herein, tremella-like CoNi-LDH intercalated by cyclic polyoxometalate (POM) anion P8W48 (P8W48/CoNi-LDH) prepared by a simple two-step hydrothermal-exfoliation assembly method is proposed as an effective electrocatalyst for NO2- to NH3 conversion. The introduction of POM with excellent redox ability tremendously increased the electrocatalytic performance of CoNi-LDH in the NO2RR process, causing P8W48/CoNi-LDH to exhibit large NH3 yield of 0.369 mmol h-1 mgcat-1 and exceptionally high Faradic efficiency of 97.0% at -1.3 V vs the Ag/AgCl reference electrode in 0.1 M phosphate buffer saline (PBS, pH = 7) containing 0.1 M NO2-. Furthermore, P8W48/CoNi-LDH demonstrated excellent durability during cyclic electrolysis. This work provides a new reference for the application of POM-based nanocomposites in the electrochemical reduction of NO2- to obtain value-added NH3.

2.
Anal Bioanal Chem ; 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38512384

RESUMEN

Electrochemiluminescence resonance energy transfer (ECL-RET) is a versatile signal transduction strategy widely used in the fabrication of chem/biosensors. However, this technique has not yet been applied in visualized imaging analysis of intracellular species due to the insulating nature of the cell membrane. Here, we construct a ratiometric ECL-RET analytical method for hypochlorite ions (ClO-) by ECL luminophore, with a luminol derivative (L-012) as the donor and a fluorescence probe (fluorescein hydrazide) as the acceptor. L-012 can emit a strong blue ECL signal and fluorescein hydrazide has negligible absorbance and fluorescence signal in the absence of ClO-. Thus, the ECL-RET process is turned off at this time. In the presence of ClO-, however, the closed-loop hydrazide structure in fluorescein hydrazide is opened via specific recognition with ClO-, accompanied with intensified absorbance and fluorescence signal. Thanks to the spectral overlap between the ECL spectrum of L-012 and the absorption spectrum of fluorescein, the ECL-RET effect is gradually recovered with the addition of ClO-. Furthermore, the ECL-RET system has been successfully applied to image intracellular ClO-. Although the insulating nature of the cell itself can generate a shadow ECL pattern in the cellular region, extracellular ECL emission penetrates the cell membrane and excites intracellular fluorescein generated by the reactions between fluorescein hydrazide and ClO-. The cell imaging strategy via ECL-RET circumvents the blocking of the cell membrane and enables assays of intracellular species. The importance of the ECL-RET platform lies in calibrating the fluctuation from the external environment and improving the selectivity by using fluorescent probes. Therefore, this ratiometric ECL sensor has shown broad application prospects in the identification of targets in clinical diagnosis and environmental monitoring.

3.
Angew Chem Int Ed Engl ; 62(30): e202305843, 2023 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-37232089

RESUMEN

The development of a new electrolytic water hydrogen production coupling system is the key to realize efficient and low-cost hydrogen production and promote its practical application. Herein, a green and efficient electrocatalytic biomass to formic acid (FA) coupled hydrogen production system has been developed. In such a system, carbohydrates such as glucose are oxidized to FA using polyoxometalates (POMs) as the redox anolyte, while H2 is evolved continuously at the cathode. Among them, the yield of glucose to FA is as high as 62.5 %, and FA is the only liquid product. Furthermore, the system requires only 1.22 V to drive a current density of 50 mA cm-2 , and the Faraday efficiency of hydrogen production is close to 100 %. Its electrical consumption is only 2.9 kWh Nm-3 (H2 ), which is only 69 % of that of traditional electrolytic water. This work opens up a promising direction for low-cost hydrogen production coupled with efficient biomass conversion.

4.
Circ Res ; 116(3): 418-24, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25351576

RESUMEN

RATIONALE: A major goal for the treatment of heart tissue damaged by cardiac injury is to develop strategies for restoring healthy heart muscle through the regeneration and repair of damaged myocardium. We recently demonstrated that administration of a specific combination of microRNAs (miR combo) into the infarcted myocardium leads to direct in vivo reprogramming of noncardiac myocytes to cardiac myocytes. However, the biological and functional consequences of such reprogramming are not yet known. OBJECTIVE: The aim of this study was to determine whether noncardiac myocytes directly reprogrammed using miRNAs in vivo develop into mature functional cardiac myocytes in situ, and whether reprogramming leads to improvement of cardiac function. METHODS AND RESULTS: We subjected fibroblast-specific protein 1-Cre mice/tandem dimer Tomato (tdTomato) mice to cardiac injury by permanent ligation of the left anterior descending coronary artery and injected lentiviruses encoding miR combo or a control nontargeting miRNA. miR combo significantly increased the number of reprogramming events in vivo. Five to 6 weeks after injury, morphological and physiological properties of tdTomato(-) and tdTomato(+) cardiac myocyte-like cells were analyzed ex vivo. tdTomato(+) cells expressed cardiac myocyte markers, sarcomeric organization, excitation-contraction coupling, and action potentials characteristic of mature ventricular cardiac myocytes (tdTomato(-) cells). Reprogramming was associated with improvement of cardiac function, as analyzed by serial echocardiography. There was a time delayed and progressive improvement in fractional shortening and other measures of ventricular function, indicating that miR combo promotes functional recovery of damaged myocardium. CONCLUSIONS: The findings from this study further validate the potential use of miRNA-mediated reprogramming as a therapeutic approach to promote cardiac regeneration after myocardial injury.


Asunto(s)
Reprogramación Celular , MicroARNs/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/citología , Animales , Fibroblastos/citología , Fibroblastos/metabolismo , Regeneración Tisular Dirigida , Masculino , Ratones , MicroARNs/genética , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Proteína de Unión al Calcio S100A4 , Proteínas S100/genética , Proteínas S100/metabolismo
5.
Circ Res ; 115(12): 1007-16, 2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25296984

RESUMEN

RATIONALE: Cardiac progenitor cells (CPCs) are thought to differentiate into the major cell types of the heart: cardiomyocytes, smooth muscle cells, and endothelial cells. We have recently identified ABI family, member 3 (NESH) binding protein (Abi3bp) as a protein important for mesenchymal stem cell biology. Because CPCs share several characteristics with mesenchymal stem cells, we hypothesized that Abi3bp would similarly affect CPC differentiation and proliferation. OBJECTIVE: To determine whether Abi3bp regulates CPC proliferation and differentiation. METHODS AND RESULTS: In vivo, genetic ablation of the Abi3bp gene inhibited CPC differentiation, whereas CPC number and proliferative capacity were increased. This correlated with adverse recovery after myocardial infarction. In vitro, CPCs, either isolated from Abi3bp knockout mice or expressing an Abi3bp shRNA construct, displayed a higher proliferative capacity and, under differentiating conditions, reduced expression of both early and late cardiomyocyte markers. Abi3bp controlled CPC differentiation via integrin-ß1, protein kinase C-ζ, and v-akt murine thymoma viral oncogene homolog. CONCLUSIONS: We have identified Abi3bp as a protein important for CPC differentiation and proliferation.


Asunto(s)
Proteínas Portadoras/metabolismo , Diferenciación Celular , Proliferación Celular , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Proteínas Portadoras/genética , Células Cultivadas , Modelos Animales de Enfermedad , Integrina beta1/metabolismo , Isoenzimas/metabolismo , Masculino , Células Madre Mesenquimatosas/patología , Ratones Noqueados , Contracción Miocárdica , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , Proteína Quinasa C/metabolismo , Proteína Quinasa C-theta , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Recuperación de la Función , Regeneración , Transducción de Señal , Volumen Sistólico , Factores de Tiempo , Transfección
6.
J Mol Cell Cardiol ; 85: 215-25, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26071893

RESUMEN

Wnt signaling has recently emerged as an important regulator of cardiac progenitor cell proliferation and differentiation, but the exact mechanisms by which Wnt signaling modulates these effects are not known. Understanding these mechanisms is essential for advancing our knowledge of cardiac progenitor cell biology and applying this knowledge to enhance cardiac therapy. Here, we explored the effects of Sfrp2, a canonical Wnt inhibitor, in adult cardiac progenitor cell (CPC) differentiation and investigated the molecular mechanisms involved. Our data show that Sfrp2 treatment can promote differentiation of CPCs after ischemia-reperfusion injury. Treatment of CPCs with Sfrp2 inhibited CPC proliferation and primed them for cardiac differentiation. Sfrp2 binding to Wnt6 and inhibition of Wnt6 canonical pathway was essential for the inhibition of CPC proliferation. This inhibition of Wnt6 canonical signaling by Sfrp2 was important for activation of the non-canonical Wnt/Planar Cell Polarity (PCP) pathway through JNK, which in turn induced expression of cardiac transcription factors and CPC differentiation. Taken together, these results demonstrate a novel role of Sfrp2 and Wnt6 in regulating the dynamic process of CPC proliferation and differentiation, as well as providing new insights into the mechanisms of Wnt signaling in cardiac differentiation.


Asunto(s)
Diferenciación Celular , Proteínas de la Membrana/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Células Madre/fisiología , Proteínas Wnt/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Expresión Génica , Ratones , Miocitos Cardíacos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Regulación hacia Arriba , Vía de Señalización Wnt
7.
Circ Res ; 113(4): 372-80, 2013 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-23784961

RESUMEN

RATIONALE: The regenerative capacity of the heart is markedly diminished shortly after birth, coinciding with overall withdrawal of cardiomyocytes from cell cycle. Consequently, the adult mammalian heart has limited capacity to regenerate after injury. The discovery of factors that can induce cardiomyocyte proliferation is, therefore, of high interest and has been the focus of extensive investigation throughout the past years. OBJECTIVE: We have recently identified C3orf58 as a novel hypoxia and Akt induced stem cell factor (HASF) secreted from mesenchymal stem cells, which can promote cardiac repair through cytoprotective mechanisms. Here, we tested the hypothesis that HASF can also contribute to cardiac regeneration by stimulating cardiomyocyte division and proliferation. METHODS AND RESULTS: Neonatal ventricular cardiomyocytes were stimulated in culture for 7 days with purified recombinant HASF protein. Compared with control untreated cells, HASF-treated neonatal cardiomyocytes exhibited 60% increase in DNA synthesis as measured by bromodeoxyuridine incorporation. These results were confirmed by immunofluorescence confocal microscopy showing a 50% to 100% increase in the number of cardiomyocytes in the mitotic and cytokinesis phases. Importantly, in vivo cardiac overexpression of HASF in a transgenic mouse model resulted in enhanced level of DNA synthesis and cytokinesis in neonatal and adult cardiomyocytes. These proliferative effects were modulated by a phosphoinositide 3-kinase-protein kinase B-cycle-dependent kinase 7 pathway as revealed by the use of phosphoinositide 3-kinase -pathway-specific inhibitors and silencing of the Cdk7 gene. CONCLUSIONS: Our studies support the hypothesis that HASF induces cardiomyocyte proliferation via a phosphoinositide 3-kinase-protein kinase B-cycle-dependent kinase 7 pathway. The implications of this finding may be significant for cardiac regeneration biology and therapeutics.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/farmacología , Ciclo Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/fisiología , Proteínas de la Membrana/farmacología , Miocitos Cardíacos/citología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal/fisiología , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Ciclo Celular/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , ADN/metabolismo , Corazón/fisiología , Humanos , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intercelular/farmacología , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Modelos Animales , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Ratas , Proteínas Recombinantes/farmacología , Regeneración , Transducción de Señal/efectos de los fármacos , Quinasa Activadora de Quinasas Ciclina-Dependientes
8.
Circ Res ; 110(11): 1465-73, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22539765

RESUMEN

RATIONALE: Repopulation of the injured heart with new, functional cardiomyocytes remains a daunting challenge for cardiac regenerative medicine. An ideal therapeutic approach would involve an effective method at achieving direct conversion of injured areas to functional tissue in situ. OBJECTIVE: The aim of this study was to develop a strategy that identified and evaluated the potential of specific micro (mi)RNAs capable of inducing reprogramming of cardiac fibroblasts directly to cardiomyocytes in vitro and in vivo. METHODS AND RESULTS: Using a combinatorial strategy, we identified a combination of miRNAs 1, 133, 208, and 499 capable of inducing direct cellular reprogramming of fibroblasts to cardiomyocyte-like cells in vitro. Detailed studies of the reprogrammed cells demonstrated that a single transient transfection of the miRNAs can direct a switch in cell fate as documented by expression of mature cardiomyocyte markers, sarcomeric organization, and exhibition of spontaneous calcium flux characteristic of a cardiomyocyte-like phenotype. Interestingly, we also found that miRNA-mediated reprogramming was enhanced 10-fold on JAK inhibitor I treatment. Importantly, administration of miRNAs into ischemic mouse myocardium resulted in evidence of direct conversion of cardiac fibroblasts to cardiomyocytes in situ. Genetic tracing analysis using Fsp1Cre-traced fibroblasts from both cardiac and noncardiac cell sources strongly suggests that induced cells are most likely of fibroblastic origin. CONCLUSIONS: The findings from this study provide proof-of-concept that miRNAs have the capability of directly converting fibroblasts to a cardiomyocyte-like phenotype in vitro. Also of significance is that this is the first report of direct cardiac reprogramming in vivo. Our approach may have broad and important implications for therapeutic tissue regeneration in general.


Asunto(s)
Transdiferenciación Celular , Fibroblastos/metabolismo , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Regulación de la Expresión Génica , Terapia Genética/métodos , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/administración & dosificación , Contracción Miocárdica , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/terapia , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Inhibidores de Proteínas Quinasas/farmacología , Recuperación de la Función , Regeneración , Proteínas S100/genética , Proteínas S100/metabolismo , Transfección , Proteína Fluorescente Roja
9.
J Am Soc Nephrol ; 24(8): 1263-73, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23744888

RESUMEN

The renin-angiotensin-aldosterone system (RAAS) regulates BP and salt-volume homeostasis. Juxtaglomerular (JG) cells synthesize and release renin, which is the first and rate-limiting step in the RAAS. Intense pathologic stresses cause a dramatic increase in the number of renin-producing cells in the kidney, termed JG cell recruitment, but how this occurs is not fully understood. Here, we isolated renal CD44(+) mesenchymal stem cell (MSC)-like cells and found that they differentiated into JG-like renin-expressing cells both in vitro and in vivo. Sodium depletion and captopril led to activation and differentiation of these cells into renin-expressing cells in the adult kidney. In summary, CD44(+) MSC-like cells exist in the adult kidney and can differentiate into JG-like renin-producing cells under conditions that promote JG cell recruitment.


Asunto(s)
Células Madre Adultas/metabolismo , Captopril/farmacología , Diferenciación Celular/fisiología , Aparato Yuxtaglomerular/citología , Riñón/citología , Células Madre Mesenquimatosas/metabolismo , Sistema Renina-Angiotensina/fisiología , Renina/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Aparato Yuxtaglomerular/metabolismo , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Sistema Renina-Angiotensina/efectos de los fármacos
10.
Proc Natl Acad Sci U S A ; 107(49): 21110-5, 2010 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-21078975

RESUMEN

Secreted frizzled related protein 2 (Sfrp2) is known as an inhibitor for the Wnt signaling. In recent studies, Sfrp2 has been reported to inhibit the activity of Xenopus homolog of mammalian Tolloid-like 1 metalloproteinase. Bone morphogenic protein 1 (Bmp1)/Tolloid-like metalloproteinase plays a key role in the regulation of collagen biosynthesis and maturation after tissue injury. Here, we showed both endogenous Sfrp2 and Bmp1 protein expressions were up-regulated in rat heart after myocardial infarction (MI). We hypothesize that Sfrp2 could inhibit mammalian Bmp1 activity and, hence, the exogenous administration of Sfrp2 after MI would inhibit the deposition of mature collagen and improve heart function. Using recombinant proteins, we demonstrated that Sfrp2, but not Sfrp1 or Sfrp3, inhibited Bmp1 activity in vitro as measured by a fluorogenic peptide based procollagen C-proteinase activity assay. We also demonstrated that Sfrp2 at high concentration inhibited human and rat type I procollagen processing by Bmp1 in vitro. We further showed that exogenously added Sfrp2 inhibited type I procollagen maturation in primary cardiac fibroblasts. Two days after direct injection into the rat infarcted myocardium, Sfrp2 inhibited MI-induced type I collagen deposition. As early as 2 wk after injection, Sfrp2 significantly reduced left ventricular (LV) fibrosis as shown by trichrome staining. Four weeks after injection, Sfrp2 prevented the anterior wall thinning and significantly improved cardiac function as revealed by histological analysis and echocardiographic measurement. Our study demonstrates Sfrp2 at therapeutic doses can inhibit fibrosis and improve LV function at a later stage after MI.


Asunto(s)
Fibrosis/tratamiento farmacológico , Receptores Frizzled/análisis , Receptores Frizzled/uso terapéutico , Proteínas de la Membrana/uso terapéutico , Función Ventricular Izquierda/efectos de los fármacos , Animales , Proteína Morfogenética Ósea 1/análisis , Proteína Morfogenética Ósea 1/antagonistas & inhibidores , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Receptores Frizzled/administración & dosificación , Humanos , Masculino , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/análisis , Infarto del Miocardio/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos , Disfunción Ventricular Izquierda/tratamiento farmacológico
11.
Ecotoxicol Environ Saf ; 93: 186-90, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23607973

RESUMEN

The illegal use of clenbuterol has been an increasingly serious issue in today's livestock products industry. It becomes an important project to develop a reliable approach to detect its content in food animals. A simple and sensitive LC-MS/MS method was developed to detect clenbuterol residue in hair, with the low limit of quantitation (LLOQ) about 0.5ng/g. Hogs fed with 340µg/day of clenbuterol for 2 weeks were found a high clenbuterol residue in their hair approximately at 1-2 months after withdrawal. There remained 3.31ng/g clenbuterol in hog hair approximately 5 months after the last administration, focused on the tip of the hair (mainly in hogs with dark hair). An extensive contamination was observed in twenty investigated market hogs whose dark hair obviously had a higher clenbuterol residue than the light ones (p=0.017, t test). Volunteers (60.3 percent) from Xuhui district (Shanghai) were found to have a detectable amount of clenbuterol in their hair (>0.5ng/g). In conclusion, hair residue detection is a reliable method to evaluate the clenbuterol contamination in animals and humans. Meat supply in the Xuhui district might have serious potential safety risks which should be further investigated and discussed to determine the safety range of clenbuterol residue.


Asunto(s)
Clenbuterol/análisis , Cabello/química , Agonistas Adrenérgicos beta/análisis , Agonistas Adrenérgicos beta/metabolismo , Crianza de Animales Domésticos , Animales , China , Cromatografía Liquida , Clenbuterol/metabolismo , Femenino , Contaminación de Alimentos/análisis , Cabello/metabolismo , Humanos , Ganado , Masculino , Carne/análisis , Porcinos , Espectrometría de Masas en Tándem
12.
Cell Rep ; 42(5): 112503, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37178120

RESUMEN

Striking antibody evasion by emerging circulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants drives the identification of broadly neutralizing antibodies (bNAbs). However, how a bNAb acquires increased neutralization breadth during antibody evolution is still elusive. Here, we identify a clonally related antibody family from a convalescent individual. One of the members, XG005, exhibits potent and broad neutralizing activities against SARS-CoV-2 variants, while the other members show significant reductions in neutralization breadth and potency, especially against the Omicron sublineages. Structural analysis visualizing the XG005-Omicron spike binding interface reveals how crucial somatic mutations endow XG005 with greater neutralization potency and breadth. A single administration of XG005 with extended half-life, reduced antibody-dependent enhancement (ADE) effect, and increased antibody product quality exhibits a high therapeutic efficacy in BA.2- and BA.5-challenged mice. Our results provide a natural example to show the importance of somatic hypermutation during antibody evolution for SARS-CoV-2 neutralization breadth and potency.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Ratones , Anticuerpos , Anticuerpos ampliamente neutralizantes , Mutación/genética , Anticuerpos Antivirales , Anticuerpos Neutralizantes
13.
Circ Res ; 106(11): 1753-62, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20378860

RESUMEN

RATIONALE: Although mesenchymal stem cell (MSC) transplantation has been shown to promote cardiac repair in acute myocardial injury in vivo, its overall restorative capacity appears to be restricted mainly because of poor cell viability and low engraftment in the ischemic myocardium. Specific chemokines are upregulated in the infarcted myocardium. However the expression levels of the corresponding chemokine receptors (eg, CCR1, CXCR2) in MSCs are very low. We hypothesized that this discordance may account for the poor MSC engraftment and survival. OBJECTIVE: To determine whether overexpression of CCR1 or CXCR2 chemokine receptors in MSCs augments their cell survival, migration and engraftment after injection in the infarcted myocardium. METHODS AND RESULTS: Overexpression of CCR1, but not CXCR2, dramatically increased chemokine-induced murine MSC migration and protected MSC from apoptosis in vitro. Moreover, when MSCs were injected intramyocardially one hour after coronary artery ligation, CCR1-MSCs accumulated in the infarcted myocardium at significantly higher levels than control-MSCs or CXCR2-MSCs 3 days postmyocardial infarction (MI). CCR1-MSC-injected hearts exhibited a significant reduction in infarct size, reduced cardiomyocytes apoptosis and increased capillary density in injured myocardium 3 days after MI. Furthermore, intramyocardial injection of CCR1-MSCs prevented cardiac remodeling and restored cardiac function 4 weeks after MI. CONCLUSIONS: Our results demonstrate the in vitro and in vivo salutary effects of genetic modification of stem cells. Specifically, overexpression of chemokine receptor enhances the migration, survival and engraftment of MSCs, and may provide a new therapeutic strategy for the injured myocardium.


Asunto(s)
Capilares/metabolismo , Movimiento Celular , Células Endoteliales/metabolismo , Terapia Genética/métodos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Infarto del Miocardio/terapia , Miocardio/metabolismo , Neovascularización Fisiológica , Receptores CCR1/biosíntesis , Animales , Apoptosis , Capilares/patología , Capilares/fisiopatología , Supervivencia Celular , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/patología , Femenino , Fibrosis , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Interferencia de ARN , Receptores CCR1/genética , Receptores de Interleucina-8B/biosíntesis , Receptores de Interleucina-8B/genética , Recuperación de la Función , Regeneración , Factores de Tiempo , Transducción Genética , Función Ventricular Izquierda , Remodelación Ventricular
14.
bioRxiv ; 2022 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-36561175

RESUMEN

Striking antibody evasion by emerging circulating SARS-CoV-2 variants drives the identification of broadly neutralizing antibodies (bNAbs). However, how a bNAb acquires increased neutralization breadth during antibody evolution is still elusive. Here, we identified a clonally-related antibody family from a convalescent individual. One of the members, XG005, exhibited potent and broad neutralizing activities against SARS-CoV-2 variants, while the other members showed significant reductions in neutralization breadth and potency, especially against the Omicron sublineages. Structural analysis visualizing the XG005-Omicron spike binding interface revealed how crucial somatic mutations endowed XG005 with greater neutralization potency and breadth. A single administration of XG005 with extended half-life, reduced antibody-dependent enhancement (ADE) effect, and increased antibody product quality, exhibited a high therapeutic efficacy in BA.2- and BA.5-challenged mice. Our results provided a natural example to show the importance of somatic hypermutation during antibody evolution for SARS-CoV-2 neutralization breadth and potency.

15.
J Biol Chem ; 285(16): 11974-82, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20118482

RESUMEN

Renin is a key enzyme for cardiovascular and renal homeostasis and is produced by highly specialized endocrine cells in the kidney, known as juxtaglomerular (JG) cells. The nature and origin of these cells remain as mysteries. Previously, we have shown that the nuclear hormone receptor liver X receptor-alpha (LXRalpha) is a major transcriptional regulator of the expression of renin, c-myc, and other genes involved with growth/differentiation. In this study we test the hypothesis that LXRalpha plays an important role not only in renin expression but also in renin-containing cell differentiation, specifically from the mesenchymal stem cell (MSC), which may be the origin of the JG cell. Indeed, our data demonstrated that LXRalpha activation by its ligands or cAMP stimulated renin gene expression in both murine and human MSCs. Furthermore, sustained cAMP stimulation of murine MSCs overexpressing LXRalpha led to their differentiation into JG-like cells expressing renin and alpha-smooth muscle actin. These MSC-derived JG-like cells contained renin in secretory granules and released active renin in response to cAMP. In conclusion, the activation of LXRalpha stimulates renin expression and induces MSCs differentiation into renin-secreting, JG-like cells. Our results suggest that the MSC may be the origin of the juxtaglomerular cell and provide insight into novel understanding of pathophysiology of the renin-angiotensin system.


Asunto(s)
Aparato Yuxtaglomerular/citología , Aparato Yuxtaglomerular/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Receptores Nucleares Huérfanos/metabolismo , Renina/biosíntesis , Actinas/biosíntesis , Actinas/genética , Animales , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Línea Celular , AMP Cíclico/farmacología , Expresión Génica/efectos de los fármacos , Humanos , Hidroxicolesteroles/farmacología , Técnicas In Vitro , Ligandos , Receptores X del Hígado , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Receptores Nucleares Huérfanos/antagonistas & inhibidores , Receptores Nucleares Huérfanos/genética , ARN Interferente Pequeño/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Renina/genética , Sistema Renina-Angiotensina/fisiología
16.
Stem Cells ; 27(4): 971-9, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19353525

RESUMEN

Administration of mesenchymal stem cells (MSCs) is an effective therapy to repair cardiac damage after myocardial infarction (MI) in experimental models. However, the mechanisms of action still need to be elucidated. Our group has recently suggested that MSCs mediate their therapeutic effects primarily via paracrine cytoprotective action. Furthermore, we have shown that MSCs overexpressing Akt1 (Akt-MSCs) exert even greater cytoprotection than unmodified MSCs. So far, little has been reported on the metabolic characteristics of infarcted hearts treated with stem cells. Here, we hypothesize that Akt-MSC administration may influence the metabolic processes involved in cardiac adaptation and repair after MI. MI was performed in rats randomized in four groups: sham group and animals treated with control MSCs, Akt-MSCs, or phosphate-buffered saline (PBS). High energy metabolism and basal 2-deoxy-glucose (2-DG) uptake were evaluated on isolated hearts using phosphorus-31 nuclear magnetic resonance spectroscopy at 72 hours and 2 weeks after MI. Treatment with Akt-MSCs spared phosphocreatine stores and significantly limited the increase in 2-DG uptake in the residual intact myocardium compared with the PBS- or the MSC-treated animals. Furthermore, Akt-MSC-treated hearts had normal pH, whereas low pH was measured in the PBS and MSC groups. Correlative analysis indicated that functional recovery after MI was inversely related to the rate of 2-DG uptake. We conclude that administration of MSCs overexpressing Akt at the time of infarction results in preservation of normal metabolism and pH in the surviving myocardium.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Infarto del Miocardio/metabolismo , Infarto del Miocardio/terapia , Miocardio/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Desoxiglucosa/metabolismo , Femenino , Concentración de Iones de Hidrógeno , Espectroscopía de Resonancia Magnética , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción Genética
17.
Int Immunol ; 21(7): 817-29, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19515797

RESUMEN

Myocardial ischemia with subsequent reperfusion (MI/R) can lead to significant myocardial damage. Ischemia initiates inflammation at the blood-microvascular endothelial cell interface and contributes significantly to both acute injury and repair of the damaged tissue. We have found that MI/R injury in mice is associated with a cellular immune response to troponin. Myocardial cells exclusively synthesize troponin and release the troponin into the bloodstream following injury. Mucosally administered proteins induce T cells that secrete anti-inflammatory cytokines such as IL-10 and transforming growth factor beta at the anatomical site where the protein localizes. We found that nasal administration of the three subunits of troponin (C, I and T isoforms), given prior to or 1 h following MI/R, decreased infarct size by 40% measured 24 h later. At 1.5 months following MI/R, there was a 50% reduction in infarct size and improvement in cardiac function as measured by echocardiography. Protection was associated with a reduction of cellular immunity to troponin. Immunohistochemistry demonstrated increased IL-10 and reduced IFN-gamma in the area surrounding the ischemic infarct following nasal troponin. Adoptive transfer of CD4+ T cells to mice from nasally troponin-treated mice 1 h after the MI/R decreased infarct size by 72%, whereas CD4+ T cells from IL-10-/- mice or nasally BSA-treated mice had no effect. Our results demonstrate that IL-10-secreting CD4+ T cells induced by nasal troponin reduce injury following MI/R. Modulation of cardiac inflammation by nasal troponin provides a novel treatment to decrease myocardial damage and enhance recovery after myocardial ischemia.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Interferón gamma/inmunología , Interleucina-10/inmunología , Daño por Reperfusión Miocárdica/prevención & control , Troponina/administración & dosificación , Administración Intranasal , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Interferón gamma/antagonistas & inhibidores , Interleucina-10/agonistas , Interleucina-10/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión Miocárdica/inmunología , Daño por Reperfusión Miocárdica/patología , Miocardio/inmunología , Miocardio/patología , Isoformas de Proteínas/inmunología , Troponina/inmunología , Vacunación
18.
Mol Biol Cell ; 18(8): 2873-82, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17507648

RESUMEN

Recent evidence has demonstrated the importance of bone marrow-derived mesenchymal stem cells (BM-MSCs) in the repair of damaged myocardium. The molecular mechanisms of engraftment and migration of BM-MSCs in the ischemic myocardium are unknown. In this study, we developed a functional genomics approach toward the identification of mediators of engraftment and migration of BM-MSCs within the ischemic myocardium. Our strategy involves microarray profiling (>22,000 probes) of ischemic hearts, complemented by reverse transcription-polymerase chain reaction and fluorescence-activated cell sorting of corresponding adhesion molecule and cytokine receptors in BM-MSCs to focus on the coexpressed pairs only. Our data revealed nine complementary adhesion molecules and cytokine receptors, including integrin beta1, integrin alpha4, and CXC chemokine receptor 4 (CXCR4). To examine their functional contributions, we first blocked selectively these receptors by preincubation of BM-MSCs with specific neutralizing antibodies, and then we administered these cells intramyocardially. A significant reduction in the total number of BM-MSC in the infarcted myocardium was observed after integrin beta1 blockade but not integrin alpha4 or CXCR4 blockade. The latter observation is distinctively different from that reported for hematopoietic stem cells (HSCs). Thus, our data show that BM-MSCs use a different pathway from HSCs for intramyocardial trafficking and engraftment.


Asunto(s)
Movimiento Celular , Integrina beta1/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Miocardio/patología , Receptores CXCR4/metabolismo , Animales , Anticuerpos/farmacología , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Genómica , Ligandos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Infarto del Miocardio/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Reproducibilidad de los Resultados
19.
Circ Res ; 99(3): 315-22, 2006 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-16825578

RESUMEN

Bone marrow-derived endothelial progenitor cells (EPCs) have the ability to migrate to ischemic organs. However, the signals that mediate trafficking and recruitment of these cells are not well understood. Using a functional genomics strategy, we determined the genes that were upregulated in the ischemic myocardium and might be involved in EPC recruitment. Among them, CD18 and its ligand ICAM-1 are particularly intriguing because CD18 and its heterodimer binding chains CD11a and CD11b were correspondingly expressed in ex vivo-expanded EPCs isolated from rat and murine bone marrows. To further verify the functional role of CD18 in mediating EPC recruitment and repair to the infarcted myocardium, we used neutralizing antibody to block CD18. Blockade of CD18 in EPCs significantly inhibited their attachment capacity in vitro and reduced their recruitment to the ischemic myocardium in vivo by 95%. Moreover, mice receiving EPCs that were treated with control isotype IgG exhibited significantly increased capillary density in the infarct border zone, reduced cardiac dilatation, ventricular wall thinning, and fibrosis when compared with myocardial infarction mice receiving PBS and CD18 blockade reversed the EPC-mediated improvements to the infarcted heart. Thus, our results suggest an essential role of CD18 in mediating EPC recruitment and the subsequent functional effects on the infarcted heart.


Asunto(s)
Antígenos CD18/fisiología , Movimiento Celular , Células Endoteliales/fisiología , Molécula 1 de Adhesión Intercelular/fisiología , Infarto del Miocardio , Neovascularización Fisiológica , Regeneración , Células Madre/fisiología , Animales , Células de la Médula Ósea/citología , Antígenos CD18/genética , Células Endoteliales/trasplante , Femenino , Perfilación de la Expresión Génica , Molécula 1 de Adhesión Intercelular/genética , Masculino , Ratones , Ratones Desnudos , Isquemia Miocárdica , Ratas , Ratas Sprague-Dawley , Trasplante de Células Madre , Transfección , Regulación hacia Arriba/genética
20.
Chem Asian J ; 13(2): 158-163, 2018 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-29193843

RESUMEN

A new type of P-doped Mo2 C coated by N-doped carbon (P-Mo2 C@NC) has been successfully prepared by calcining a mixture of H3 [PMo12 O40 ] polyoxometalates (POMs) and urea-formaldehyde resin under an N2 atmosphere. Urea-formaldehyde resin not only serves as the carbon source to ensure carbonization but also facilitates the uniform distribution of POM precursors, which efficiently avoid the aggregation of Mo2 C particles at high temperatures. TEM analysis revealed that the average diameter of the Mo2 C particles was about 10 nm, which is coated by a few-layer N-doped carbon sheet. The as-prepared P-Mo2 C@NC displayed excellent hydrogen-evolution reaction (HER) performance and long-term stability in all pH environments. To reach a current density of 10 mA cm-2 , only 109, 159, and 83 mV were needed for P-Mo2 C@NC in 0.5 m H2 SO4 (pH 0), 0.1 m phosphate buffer (pH 7), and 1 m KOH (pH 14), respectively. This could provide a high-yield and low-cost method to prepare uniform nanosized molybdenum carbides with highly efficient and stable HER performance.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA