Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Physiol Pharmacol ; 70(6)2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32203936

RESUMEN

Stem cell therapy in combination with genetic modification (e.g., transfection with the coding sequence for the connexion 43 gene, GJA1) may solve the problems associated with the occurrence of additional (secondary) stimulation in the post-infarcted heart (arrhythmia). Human skeletal muscle-derived stem/progenitor cells (SkMDS/PCs) were transfected with the pCiNeo-GJA1 plasmid at an efficiency of approximately 96%. Gene overexpression was assessed using qPCR, and subsequent analysis revealed that GJA1 expression increased more than 40-fold in SkMDS/PCs transfected with the appropriate coding sequence (SkMDS/PCsCX43) compared to that of the 'native' SkMDS/PCs control (SkMDS/PCsWT). Enhanced (4-fold) protein expression of connexin-43 was also confirmed by Western immunoblotting. Furthermore, using the arrhythmic score, we demonstrated the positive effects of SkMDS/PCsCX43 cell intervention in reducing additional secondary stimulations in rat post-infarcted hearts compared with that of wild-type cell delivery. Selected gene responses (Kcnq1, Cacna1c, Ncx1, Serca2a, and Tgfb1) showed significantly altered expression profiles in the rat myocardium upon intervention with SkMDS/PCsCX43. The genetic modification of human skeletal muscle-derived stem/progenitor cells with connexin-43 prevented the pro-arrhythmic effects of myogenic implanted stem cells on the host myocardium and positively influenced myocardial gene expression profiles in respect to myocardium conductivity.


Asunto(s)
Arritmias Cardíacas/prevención & control , Conexina 43/metabolismo , Infarto del Miocardio/terapia , Trasplante de Células Madre/métodos , Animales , Arritmias Cardíacas/etiología , Conexina 43/genética , Femenino , Regulación de la Expresión Génica , Humanos , Músculo Esquelético/citología , Infarto del Miocardio/complicaciones , Miocardio/metabolismo , Ratas , Ratas Wistar , Células Madre/citología , Transfección
2.
J Physiol Pharmacol ; 69(6)2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30898982

RESUMEN

Cardiovascular diseases along with MI (myocardial infarction) lead to regional ischaemia and hypoxic conditions, which prevail after infarction. Diminished O2 saturation which is related to elevated level of hypoxia inducible factor 1 (HIF-1) transcription factor, may switch the expression of many genes. To maximize effect of therapies proposed by regenerative medicine, it is essential to verify (within different time points after MI) the expression of proangiogenic genes and their receptors that are regulated, along with the expression of HIF-1α. We demonstrated a connection between the expression of Hif-1α (in murine post infarcted heart model) and the proangiogenic genes Vegf-a; and Plgf and their receptors during myocardial hypoxia. The innovative part of the study required establishment of the most accurate in vitro O2 level corresponding to the hypoxia level prevailing in myocardium after MI. We determined the influence of hypoxia on the biology of human myoblasts in in vitro oxygen conditions (3%), corresponding to those prevailing in the heart after an infarction using a murine model. We also tested myoblasts that were genetically modified with VEGF-A/FGF-4 and PlGF under hypoxic conditions and compared their characteristics with cells cultured under normoxia and hyperoxia (standard in vitro conditions) with respect to myogenic gene expression, cell proliferation, fusion potential and proangiogenic function. The examination of genetically modified myoblasts under optimized in vitro hypoxia conditions led to the conclusion that hypoxia did not negatively influence the biological functions of the myoblasts, such as cell proliferation and/or proangiogenic characteristics. These results support the expected increased proregenerative effects of such genetically modified human myoblasts.


Asunto(s)
Expresión Génica/genética , Hipoxia/genética , Mioblastos/patología , Infarto del Miocardio/genética , Neovascularización Patológica/genética , Animales , Línea Celular , Proliferación Celular/genética , Modelos Animales de Enfermedad , Factor 4 de Crecimiento de Fibroblastos/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Ratones SCID , Miocardio/patología , Factor de Crecimiento Placentario/genética , Factor A de Crecimiento Endotelial Vascular/genética
3.
Int J Cardiol ; 202: 710-21, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26457413

RESUMEN

BACKGROUND: Myocardial infarction (MI) and left ventricle remodeling (LVR) are two of the most challenging disease entities in developed societies. Since conventional treatment cannot fully restore heart function new approaches were attempted to develop new strategies and technologies that could be used for myocardial regeneration. One of these strategies pursued was a cell therapy--particularly applying skeletal muscle stem cells (SkMCs). METHODS AND RESULTS: Using NOD-SCID murine model of MI and human skeletal myoblast transplantation we were able to show that SkMC administration significantly affected gene expression profile (p<0.05) (NPPB, CTGF, GATA4, SERCA2a, PLB) of the heart ventricular tissue and this change was beneficial for the heart function. We have also shown, that the level of heart biomarker, NT-proBNP, decreased in animals receiving implanted cells and that the NT-proBNP level negatively correlated with left ventricle area fraction change (LVFAC) index which makes NT-proBNP an attractive tool in assessing the efficacy of cell therapy both in the animal model and prospectively in clinical trials. CONCLUSIONS: The results obtained suggest that transplanted SkMCs exerted beneficial effect on heart regeneration and were able to inhibit LVR which was confirmed on the molecular level, giving hope for new ways of monitoring novel cellular therapies for MI.


Asunto(s)
Trasplante de Células/métodos , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Mioblastos/trasplante , Infarto del Miocardio/cirugía , ARN/genética , Remodelación Ventricular/fisiología , Animales , Ligamento Cruzado Anterior/citología , Células Cultivadas , Modelos Animales de Enfermedad , Citometría de Flujo , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos NOD , Ratones SCID , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocardio/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Función Ventricular Izquierda/fisiología
5.
Tech Coloproctol ; 19(11): 685-96, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26266767

RESUMEN

BACKGROUND: The aim of this study is to present results of the implantation of autologous myoblasts into the external anal sphincter (EAS) in ten patients with fecal incontinence. METHODS: After anatomical and functional assessment of the patients' EAS, a vastus lateralis muscle open biopsy was performed. Stem cells were extracted from the biopsy specimens and cultured in vitro. Cell suspensions were then administered to the EAS. Patients were scheduled for follow-up visits in 6-week intervals. Total follow-up was 12 months. RESULTS: All biopsy and cell implantation procedures were performed without complications. Nine of the patients completed a full 12-month follow-up. There was subjective improvement in six patients (66.7 %). In manometric examinations 18 weeks after implantation, squeeze anal pressures and high-pressure zone length increased in all patients, with particularly significant sphincter function recovery in five patients (55.6 %). Electromyographic (EMG) examination showed an increase in signal amplitude in all patients, detecting elevated numbers of propagating action potentials. Twelve months after implantation two patients experienced deterioration of continence, which was also reflected in the deterioration of manometric and EMG parameters. The remaining four patients (44.4 %) still described their continence as better than before implantation and retained satisfactory functional examination parameters. CONCLUSIONS: Implantation of autologous myoblasts gives good short-term results not only in a subjective assessment, but also in objective functional tests. It seems that this promising technology can improve the quality of life of patients with fecal incontinence, but further study is required to achieve better and more persistent results.


Asunto(s)
Canal Anal , Incontinencia Fecal/cirugía , Mioblastos/trasplante , Recuperación de la Función , Adulto , Anciano , Canal Anal/fisiopatología , Canal Anal/cirugía , Electromiografía , Incontinencia Fecal/fisiopatología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Manometría , Persona de Mediana Edad , Proyectos Piloto , Presión , Estudios Prospectivos , Músculo Cuádriceps/citología , Músculo Cuádriceps/cirugía , Trasplante Autólogo/métodos , Resultado del Tratamiento , Adulto Joven
6.
Int J Cardiol ; 173(1): 55-64, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24636551

RESUMEN

BACKGROUND: Previously, connexin 43-modified skeletal myoblasts (MbCx) were shown to reduce the pro-arrhythmic effect during the regeneration of heart tissue in an animal model of infarction. To increase the relevance to clinical implementation, in this study, we introduced connexin 43 into human myoblasts using a highly safe non-viral vector and demonstrated that their transplantation had a positive effect on the function of the injured heart. METHODS AND RESULTS: Myoblasts were efficiently transfected with a pCiNeo-GJA1 plasmid (65.72%). qPCR analysis revealed over 32-fold higher expression of the connexin 43 gene in the MbCx cell population compared to 'native' controls. The susceptibility of the myoblasts to oxidative stress conditions (p<0.001) and the fusion index (p<0.01) were increased in the MbCx cells. Additionally, we observed changes in the MYOG and MYH2 gene expression levels in the GJA1-modified myoblasts. Finally, we observed a significant improvement in the post-infarction echocardiographic parameters after intervention using MbCx cells compared with non-transfected myoblasts (MbWt) and the control (0.9% NaCl), wherein a significant decrease in the left ventricular area change in the short axis (SAX AC%) was observed at the two-month follow-up (p<0.05 and p<0.01, respectively). CONCLUSIONS: We demonstrated the positive effect of connexin 43 overexpression on the biology and function of human skeletal myoblasts in the context of their potential clinical applications. Our preclinical studies using a mouse infarction model indicated the positive effect of MbCx implantation on the function of the injured heart.


Asunto(s)
Conexina 43/biosíntesis , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/terapia , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/trasplante , Trasplante de Células Madre/métodos , Animales , Células Cultivadas , Insuficiencia Cardíaca/patología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Estudios Prospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA