Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Emerg Med ; 76: 105-110, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38056055

RESUMEN

BACKGROUND: The optimal management of patients taking oral anticoagulants who experience minor head injury (MHI) is unclear. The availability of validated protocols and reliable predictors of prognosis would be of great benefit. We investigated clinical factors as predictors of clinical outcomes and intracranial injury (ICI). METHODS: We conducted a single-cohort, prospective, observational study in an ED. Our structured clinical pathway included a first head CT scan, 24 h observation and a second CT scan. The primary outcome was the occurrence of MHI-related death or re-admission to ED at day +30. The secondary outcome was the rate of delayed ICI (dICI), defined as second positive CT scan after a first negative CT scan. We assessed some clinical predictors derived from guidelines and clinical prediction rules as potential risk factors for the outcomes. RESULTS: 450 patients with a negative first CT scan who underwent a second CT scan composed our 'study population'. The rate of the primary outcome was 4%. The rate of the secondary outcome was 4.7%. Upon univariate and multivariate analysis no statistically significant predictors for the outcomes were found. CONCLUSIONS: Previous retrospective studies showed a lot of negative predictive factors for anticoagulated patients suffering a minor head injury. In our prospective study no clinical factors emerged as predictors of poor clinical outcomes and dICI. So, even if we confirmed a low rate of adverse outcomes, the best management of these patients in ED remains not so clear and future trials are needed.


Asunto(s)
Traumatismos Craneocerebrales , Humanos , Estudios Prospectivos , Traumatismos Craneocerebrales/complicaciones , Traumatismos Craneocerebrales/diagnóstico por imagen , Anticoagulantes/efectos adversos , Factores de Riesgo , Estudios Retrospectivos
2.
Differentiation ; 133: 25-39, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37451110

RESUMEN

Cerebral palsy (CP) is one of the most common conditions leading to lifelong childhood physical disability. Literature reported previously altered muscle properties such as lower number of satellite cells (SCs), with altered fusion capacity. However, these observations highly vary among studies, possibly due to heterogeneity in patient population, lack of appropriate control data, methodology and different assessed muscle. In this study we aimed to strengthen previous observations and to understand the heterogeneity of CP muscle pathology. Myogenic differentiation of SCs from the Medial Gastrocnemius (MG) muscle of patients with CP (n = 16, 3-9 years old) showed higher fusion capacity compared to age-matched typically developing children (TD, n = 13). Furthermore, we uniquely assessed cells of two different lower limb muscles and showed a decreased myogenic potency in cells from the Semitendinosus (ST) compared to the MG (TD: n = 3, CP: n = 6). Longitudinal assessments, one year after the first botulinum toxin treatment, showed slightly reduced SC representations and lower fusion capacity (n = 4). Finally, we proved the robustness of our data, by assessing in parallel the myogenic capacity of two samples from the same TD muscle. In conclusion, these data confirmed previous findings of increased SC fusion capacity from MG muscle of young patients with CP compared to age-matched TD. Further elaboration is reported on potential factors contributing to heterogeneity, such as assessed muscle, CP progression and reliability of primary outcome parameters.


Asunto(s)
Células Madre Adultas , Parálisis Cerebral , Contractura , Humanos , Niño , Preescolar , Parálisis Cerebral/patología , Reproducibilidad de los Resultados , Músculo Esquelético/patología , Contractura/patología
3.
J Nutr Health Aging ; 23(8): 739-745, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31560032

RESUMEN

OBJECTIVES: Autophagy is a physiological and highly regulated mechanism, crucial for cell homeostasis maintenance. Its impairment seems to be involved in the onset of several diseases, including muscular dystrophies, myopathies and sarcopenia. According to few papers, chemotherapeutic drug treatment is able to trigger side effects on skeletal muscle tissue and, among these, a defective autophagic activation, which leads to the persistence of abnormal organelles within cells and, finally, to myofiber degeneration. The aim of this work is to find a strategy, based on diet modulation, to prevent etoposide-induced damage, in a model of in vitro skeletal muscle cells. METHODS: Glutamine supplementation and nutrient deprivation have been chosen as pre-treatments to counteract etoposide effect, a chemotherapeutic drug known to induce oxidative stress and cell death. Cell response has been evaluated by means of morpho-functional, cytofluorimetric and molecular analyses. RESULTS: Etoposide treated cells, if compared to control, showed dysfunctional mitochondria presence, ER stress and lysosomal compartment damage, confirmed by molecular investigations. CONCLUSIONS: Interestingly, both dietary approaches were able to rescue myofiber from etoposide-induced damage. Glutamine supplementation, in particular, seemed to be a good strategy to preserve cell ultrastructure and functionality, by preventing the autophagic impairment and partially restoring the normal lysosomal activity, thus maintaining skeletal muscle homeostasis.


Asunto(s)
Autofagia/fisiología , Dieta/métodos , Músculo Esquelético/fisiopatología , Humanos
4.
Nat Commun ; 10(1): 492, 2019 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-30700722

RESUMEN

There is an urgent need to develop the next-generation vectors for gene therapy of muscle disorders, given the relatively modest advances in clinical trials. These vectors should express substantially higher levels of the therapeutic transgene, enabling the use of lower and safer vector doses. In the current study, we identify potent muscle-specific transcriptional cis-regulatory modules (CRMs), containing clusters of transcription factor binding sites, using a genome-wide data-mining strategy. These novel muscle-specific CRMs result in a substantial increase in muscle-specific gene transcription (up to 400-fold) when delivered using adeno-associated viral vectors in mice. Significantly higher and sustained human micro-dystrophin and follistatin expression levels are attained than when conventional promoters are used. This results in robust phenotypic correction in dystrophic mice, without triggering apoptosis or evoking an immune response. This multidisciplinary approach has potentially broad implications for augmenting the efficacy and safety of muscle-directed gene therapy.


Asunto(s)
Biología Computacional/métodos , Terapia Genética/métodos , Músculo Esquelético/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Vectores Genéticos/genética , Humanos , Masculino , Ratones , Ratones SCID , Mutación/genética , Regiones Promotoras Genéticas/genética
5.
Biotech Histochem ; 90(6): 424-31, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25893542

RESUMEN

Novel clearing techniques have revolutionized three-dimensional confocal imaging of the brain without the need for physical tissue sectioning. We evaluated three clearing methods, ScaleA2, Clear(T2), and 3DISCO for visualizing native and tissue engineered muscle by confocal microscopy. We found that Clear(T2) treatment improved the depth of visualization of immunohistochemical staining slightly, but did not improve depth of visualization of endogenous green fluorescent protein (GFP). ScaleA2 preserved endogenous GFP signal better and permitted significantly deeper GFP imaging, but it was incompatible with tropomyosin immunohistochemical staining. 3DISCO treatment preserved both endogenous GFP and immunohistochemical staining, and permitted significantly deeper imaging. Clearing time for the 3DISCO procedure is short compared to ScaleA2 and Clear(T2). We suggest that 3DISCO is the preferable clearing method for native and tissue engineered skeletal muscle tissue.


Asunto(s)
Órganos Bioartificiales , Microscopía Confocal/métodos , Músculo Esquelético/anatomía & histología , Músculo Esquelético/metabolismo , Animales , Células Cultivadas , Proteínas Fluorescentes Verdes/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Imagenología Tridimensional , Inmunohistoquímica , Ratones , Ratones Transgénicos , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/metabolismo , Proteínas Recombinantes/metabolismo , Ingeniería de Tejidos
6.
Cell Death Dis ; 5: e1448, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25299773

RESUMEN

Somatic stem cells hold attractive potential for the treatment of muscular dystrophies (MDs). Mesoangioblasts (MABs) constitute a myogenic subset of muscle pericytes and have been shown to efficiently regenerate dystrophic muscles in mice and dogs. In addition, HLA-matched MABs are currently being tested in a phase 1 clinical study on Duchenne MD patients (EudraCT #2011-000176-33). Many reports indicate that the Notch pathway regulates muscle regeneration and satellite cell commitment. However, little is known about Notch-mediated effects on other resident myogenic cells. To possibly potentiate MAB-driven regeneration in vivo, we asked whether Notch signaling played a pivotal role in regulating MAB myogenic capacity. Through different approaches of loss- and gain-of-function in murine and human MABs, we determined that the interplay between Delta-like ligand 1 (Dll1)-activated Notch1 and Mef2C supports MAB commitment in vitro and ameliorates engraftment and functional outcome after intra-arterial delivery in dystrophic mice. Furthermore, using a transgenic mouse model of conditional Dll1 deletion, we demonstrated that Dll1 ablation, either on the injected cells, or on the receiving muscle fibers, impairs MAB regenerative potential. Our data corroborate the perspective of advanced combinations of cell therapy and signaling tuning to enhance therapeutic efficaciousness of somatic stem cells.


Asunto(s)
Desarrollo de Músculos , Receptor Notch1/metabolismo , Transducción de Señal , Células Madre/citología , Animales , Proteínas de Unión al Calcio , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/metabolismo , Receptor Notch1/genética , Células Madre/metabolismo
7.
Histochem Cell Biol ; 139(1): 35-46, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22936275

RESUMEN

Protein kinase Cepsilon (PKCε) exerts a well-known cardio-protective activity in ischemia-reperfusion injury and plays a pivotal role in stem cell proliferation and differentiation. Although many studies have been performed on physiological and morphological effects of PKCε mis-expression in cardiomyocytes, molecular information on the role of PKCε on early cardiac gene expression are still lacking. We addressed the molecular role of PKCε in cardiac cells using mouse cardiomyocytes and rat bone marrow mesenchymal stem cells. We show that PKCε is modulated in cardiac differentiation producing an opposite regulation of the cardiac genes NK2 transcription factor related, locus 5 (nkx2.5) and GATA binding protein 4 (gata4) both in vivo and in vitro. Phospho-extracellular regulated mitogen-activated protein kinase 1/2 (p-ERK1/2) levels increase in PKCε over-expressing cells, while pkcε siRNAs produce a decrease in p-ERK1/2. Indeed, pharmacological inhibition of ERK1/2 rescues the expression levels of both nkx2.5 and gata4, suggesting that a reinforced (mitogen-activated protein kinase) MAPK signaling is at the basis of the observed inhibition of cardiac gene expression in the PKCε over-expressing hearts. We demonstrate that PKCε is critical for cardiac cell early gene expression evidencing that this protein is a regulator that has to be fine tuned in precursor cardiac cells.


Asunto(s)
Diferenciación Celular , Células Madre Mesenquimatosas/enzimología , Miocitos Cardíacos/enzimología , Proteína Quinasa C-epsilon/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Factor de Transcripción GATA4/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Proteína Quinasa C-epsilon/genética , Interferencia de ARN , Ratas , Ratas Wistar , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección
8.
Cell Transplant ; 21(9): 1945-67, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22513051

RESUMEN

Among the animal models of Duchenne muscular dystrophy (DMD), the Golden Retriever muscular dystrophy (GRMD) dog is considered the best model in terms of size and pathological onset of the disease. As in human patients presenting with DMD or Becker muscular dystrophies (BMD), the GRMD is related to a spontaneous X-linked mutation of dystrophin and is characterized by myocardial lesions. In this respect, GRMD is a useful model to explore cardiac pathogenesis and for the development of therapeutic protocols. To investigate whether cardiac progenitor cells (CPCs) isolated from healthy and GRMD dogs may differentiate into myocardial cell types and to test the feasibility of cell therapy for cardiomyopathies in a preclinical model of DMD, CPCs were isolated from cardiac biopsies of healthy and GRMD dogs. Gene profile analysis revealed an active cardiac transcription network in both healthy and GRMD CPCs. However, GRMD CPCs showed impaired self-renewal and cardiac differentiation. Population doubling and telomerase analyses highlighted earlier senescence and proliferation impairment in progenitors isolated from GRMD cardiac biopsies. Immunofluorescence analysis revealed that only wt CPCs showed efficient although not terminal cardiac differentiation, consistent with the upregulation of cardiac-specific proteins and microRNAs. Thus, the pathological condition adversely influences the cardiomyogenic differentiation potential of cardiac progenitors. Using PiggyBac transposon technology we marked CPCs for nuclear dsRed expression, providing a stable nonviral gene marking method for in vivo tracing of CPCs. Xenotransplantation experiments in neonatal immunodeficient mice revealed a valuable contribution of CPCs to cardiomyogenesis with homing differences between wt and dystrophic progenitors. These results suggest that cardiac degeneration in dystrophinopathies may account for the progressive exhaustion of local cardiac progenitors and shed light on cardiac stemness in physiological and pathological conditions. Furthermore, we provide essential information that canine CPCs may be used to alleviate cardiac involvement in a large preclinical model of DMD.


Asunto(s)
Distrofia Muscular Animal/patología , Miocardio/citología , Miocardio/patología , Células Madre/patología , Animales , Diferenciación Celular/fisiología , Linaje de la Célula , Modelos Animales de Enfermedad , Perros , Femenino , Citometría de Flujo , Humanos , Masculino , Ratones , Ratones SCID , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Miocardio/metabolismo , Ratas , Células Madre/metabolismo , Transcriptoma
9.
Res Vet Sci ; 91(1): 18-24, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20732703

RESUMEN

Adult stem cells are nowadays used for treating several pathologies. A putative stem cell population was found in the adipose tissue of mammals and canine adipose tissue-derived-mesenchymal stem cells (cA-MSC) have been shown to possess the capacity to differentiate into several lineages. The main goal of our research was to fully characterize cA-MSC and examine the effects of cryopreservation on their stemness features. Each sample of cA-MSC was analyzed immediately and then again after being frozen in liquid nitrogen for one year. After the cryopreservation period cells conserved their fibroblast-like morphology, alkaline phosphatase positivity and CD expression but showed a lower proliferation ratio and a lower telomerase activity in comparison with fresh cells. Finally, the cryopreservation protocol did not change the cA-MSC adipogenic, osteogenic and myogenic differentiative potential. Our data demonstrate that stored cA-MSC might represent a promising type of progenitor cell for autologous cellular-based therapies in veterinary medicine.


Asunto(s)
Tejido Adiposo/citología , Criopreservación/veterinaria , Células Madre Mesenquimatosas/fisiología , Fosfatasa Alcalina/metabolismo , Animales , Antígenos de Superficie/metabolismo , Técnicas de Cultivo de Célula/veterinaria , Diferenciación Celular , Perros , Femenino , Citometría de Flujo/veterinaria , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Reacción en Cadena de la Polimerasa/veterinaria , Análisis de Secuencia de ARN/veterinaria , Telomerasa/metabolismo
10.
Cell Death Differ ; 17(8): 1222-9, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19876070

RESUMEN

Understanding stem cell commitment and differentiation is a critical step towards clinical translation of cell therapies. In past few years, several cell types have been characterized and transplanted in animal models for different diseased tissues, eligible for a cell-mediated regeneration. Skeletal muscle damage is a challenge for cell- and gene-based therapeutical approaches, given the unique architecture of the tissue and the clinical relevance of acute damages or dystrophies. In this review, we will consider the regenerative potential of embryonic and somatic stem cells and the outcomes achieved on their transplantation into animal models for muscular dystrophy or acute muscle impairment.


Asunto(s)
Distrofias Musculares/terapia , Trasplante de Células Madre , Animales , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Células Madre Embrionarias/trasplante , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología
11.
Cell Death Differ ; 15(9): 1417-28, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18497758

RESUMEN

Different cardiac stem/progenitor cells have been recently identified in the post-natal heart. We describe here the identification, clonal expansion and characterization of self-renewing progenitors that differ from those previously described for high spontaneous cardiac differentiation. Unique coexpression of endothelial and pericyte markers identify these cells as cardiac mesoangioblasts and allow prospective isolation and clonal expansion from the juvenile mouse ventricle. Cardiac mesoangioblasts express many cardiac transcription factors and spontaneously differentiate into beating cardiomyocytes that assemble mature sarcomeres and express typical cardiac ion channels. Cells similarly isolated from the atrium do not spontaneously differentiate. When injected into the ventricle after coronary artery ligation, cardiac mesoangioblasts efficiently generate new myocardium in the peripheral area of the necrotic zone, as they do when grafted in the embryonic chick heart. These data identify cardiac mesoangioblasts as committed progenitors, downstream of earlier stem/progenitor cells and suitable for the cell therapy of a subset of juvenile cardiac diseases.


Asunto(s)
Ventrículos Cardíacos/citología , Miocitos Cardíacos/citología , Células Madre/citología , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Proliferación Celular , Endotelio Vascular/citología , Ventrículos Cardíacos/crecimiento & desarrollo , Humanos , Ratones , Miocardio/citología , Técnicas de Placa-Clamp , Ratas , Células Madre/metabolismo , Células Madre/fisiología
12.
J Biomed Mater Res A ; 84(4): 1094-101, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17685407

RESUMEN

Skeletal myogenesis is a complex process, which is known to be intimately depending on an optimal outside-in substrate-cell signaling. Current attempts to reproduce skeletal muscle tissue in vitro using traditional scaffolds mainly suffer from poor directionality of the myofibers, resulting in an ineffective vectorial power generation. In this study, we aimed at investigating skeletal myogenesis on novel biodegradable microfibrous scaffolds made of DegraPol, a block polyesterurethane previously demonstrated to be suitable for this application. DegraPol was processed by electrospinning in the form of highly orientated ("O") and nonorientated ("N/O") microfibrous meshes and by solvent-casting in the form of nonporous films ("F"). The effect of the fiber orientation at the scaffold surface was evaluated by investigating C2C12 and L6 proliferation (via SEM analysis and alamarBlue test) and differentiation (via RT-PCR analysis and MHC immunostaining). We demonstrated that highly orientated elastomeric microfibrous DegraPol scaffolds enable skeletal myogenesis in vitro by aiding in (a) myoblast adhesion, (b) myotube alignment, and (c) noncoplanar arrangement of cells, by providing the necessary directional cues along with architectural and mechanical support.


Asunto(s)
Materiales Biocompatibles/química , Desarrollo de Músculos , Poliésteres/química , Poliuretanos/química , Animales , Adhesión Celular , Línea Celular , Diseño de Equipo , Ensayo de Materiales , Ratones , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Ratas , Ingeniería de Tejidos/métodos
13.
Cell Transplant ; 16(6): 563-77, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17912948

RESUMEN

Duchenne muscular dystrophy (DMD) is a lethal X-linked recessive muscle disease due to defect on the gene encoding dystrophin. The lack of a functional dystrophin in muscles results in the fragility of the muscle fiber membrane with progressive muscle weakness and premature death. There is no cure for DMD and current treatment options focus primarily on respiratory assistance, comfort care, and delaying the loss of ambulation. Recent works support the idea that stem cells can contribute to muscle repair as well as to replenishment of the satellite cell pool. Here we tested the safety of autologous transplantation of muscle-derived CD133+ cells in eight boys with Duchenne muscular dystrophy in a 7-month, double-blind phase I clinical trial. Stem cell safety was tested by measuring muscle strength and evaluating muscle structures with MRI and histological analysis. Timed cardiac and pulmonary function tests were secondary outcome measures. No local or systemic side effects were observed in all treated DMD patients. Treated patients had an increased ratio of capillary per muscle fibers with a switch from slow to fast myosin-positive myofibers.


Asunto(s)
Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Distrofia Muscular de Duchenne/terapia , Mioblastos Esqueléticos/trasplante , Péptidos/metabolismo , Antígeno AC133 , Adolescente , Antígenos CD/clasificación , Antígenos CD/aislamiento & purificación , Niño , Método Doble Ciego , Estudios de Factibilidad , Estudios de Seguimiento , Glicoproteínas/clasificación , Glicoproteínas/aislamiento & purificación , Humanos , Separación Inmunomagnética/clasificación , Inmunofenotipificación/clasificación , Inyecciones Intramusculares , Masculino , Contracción Muscular/fisiología , Músculo Esquelético/citología , Distrofia Muscular de Duchenne/patología , Mioblastos Esqueléticos/citología , Péptidos/clasificación , Péptidos/aislamiento & purificación , Trasplante de Células Madre , Células Madre/citología , Trasplante Autólogo , Trasplante Homólogo/efectos adversos , Resultado del Tratamiento
14.
FASEB J ; 21(13): 3573-83, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17601985

RESUMEN

Satellite cells are the main source of myogenic progenitors in postnatal skeletal muscle, but their use in cell therapy for muscle disorders is limited because these cells cannot be delivered through circulation and they are rapidly exhausted in severe myopathies. The search for alternative donor cells is ongoing, but none of the candidates so far show all the features required for successful colonization and repair of diseased muscle. In this study, we show that bisperoxovanadium, a phospho-tyrosine phosphatase inhibitor, induces myogenic cells to acquire a gene expression profile and a differentiation potential consistent with the phenotype of a circulating precursors, while maintaining their myogenic potential. These effects are mediated, at least in part, by NF-kappaB activation through the Tyr42-IkappaB-alpha phosphorylation, as shown by the expression of the dominant negative mutant form of the p50 NF-kappaB subunit. Moreover, when bisperoxovanadium-treated cells are injected into the femoral artery of alpha-sarcoglican null dystrophic mice, they are able to circulate and to reach muscle tissue; importantly, they contribute to muscle regeneration, as shown by the expression of alpha-sarcoglican in some fibers. Our observations indicate that bisperoxovanadium, or similar compounds, may prove very valuable to obtain and to expand, from committed cells, multipotent cell populations suitable for gene-cell therapy applications and may help to understand the molecular basis of genome reprogramming and "stem-ness."


Asunto(s)
Inhibidores Enzimáticos/farmacología , Corazón/efectos de los fármacos , Miocardio/citología , Células Madre Pluripotentes/citología , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Compuestos de Vanadio/farmacología , Animales , Secuencia de Bases , Ciclo Celular , Línea Celular , Cartilla de ADN , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Expresión Génica , Ratones , Miocardio/metabolismo , Fenotipo , Células Madre Pluripotentes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Cell Death Differ ; 14(6): 1162-71, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17347663

RESUMEN

Stromal stem cells from human dental pulp (SBP-DPSCs) were used to study osteogenic differentiation in vitro and in vivo. We previously reported that SBP-DPSCs are multipotent stem cells able to differentiate into osteoblasts, which synthesize three-dimensional woven bone tissue chips in vitro. In this study, we followed the temporal expression pattern of specific markers in SBP-DPSCs and found that, when differentiating into osteoblasts, they express, besides osteocalcin, also flk-1 (VEGF-R2). In addition, 30% of them expressed specific antigens for endothelial cells, including CD54, von-Willebrand (domain 1 and 2), CD31 (PECAM-1) and angiotensin-converting enzyme. Interestingly, we found endotheliocytes forming vessel walls, observing that stem cells synergically differentiate into osteoblasts and endotheliocytes, and that flk-1 exerts a pivotal role in coupling osteoblast and endotheliocyte differentiation. When either SBP-DPSCs or bone chips obtained in vitro were transplanted into immunocompromised rats, they generated a tissue structure with an integral blood supply similar to that of human adult bone; in fact, a large number of HLA-1+ vessels were observed either within the bone or surrounding it in a periosteal layer. This study provides direct evidence to suggest that osteogenesis and angiogenesis mediated by human SBP-DPSCs may be regulated by distinct mechanisms, leading to the organization of adult bone tissue after stem cell transplantation.


Asunto(s)
Diferenciación Celular , Pulpa Dental/citología , Osteoblastos/citología , Osteogénesis , Adipocitos/citología , Adipocitos/metabolismo , Adulto , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Pulpa Dental/metabolismo , Pulpa Dental/ultraestructura , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Microscopía Electrónica de Transmisión , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Neuronas/citología , Neuronas/metabolismo , Osteoblastos/metabolismo , Osteoblastos/ultraestructura , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante de Células Madre/métodos , Células Madre/citología , Células Madre/metabolismo , Células Madre/ultraestructura , Células del Estroma/citología , Células del Estroma/metabolismo , Células del Estroma/ultraestructura , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Factores de Tiempo
16.
Nat Med ; 12(10): 1147-50, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16980968

RESUMEN

Pharmacological interventions that increase myofiber size counter the functional decline of dystrophic muscles. We show that deacetylase inhibitors increase the size of myofibers in dystrophin-deficient (MDX) and alpha-sarcoglycan (alpha-SG)-deficient mice by inducing the expression of the myostatin antagonist follistatin in satellite cells. Deacetylase inhibitor treatment conferred on dystrophic muscles resistance to contraction-coupled degeneration and alleviated both morphological and functional consequences of the primary genetic defect. These results provide a rationale for using deacetylase inhibitors in the pharmacological therapy of muscular dystrophies.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Músculos/enzimología , Músculos/patología , Distrofia Muscular Animal/tratamiento farmacológico , Animales , Distrofina/genética , Fibrosis/patología , Folistatina/metabolismo , Ácidos Hidroxámicos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculos/efectos de los fármacos , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patología , Fenilbutiratos/farmacología , Sarcoglicanos/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/enzimología , Ácido Valproico/farmacología
17.
Arch Ital Biol ; 143(3-4): 235-42, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16097501

RESUMEN

Mesoangioblasts are multipotent progenitors of mesodermal tissues. In vitro mesoangioblasts differentiate into many mesoderm cell types, such as smooth, cardiac and striated muscle, bone and endothelium. After transplantation mesoangioblasts colonize mostly mesoderm tissues and differentiate into many cell types of the mesoderm. When delivered through the arterial circulation, mesoangioblasts significantly restore skeletal muscle structure and function in a mouse model of muscular dystrophy. Their ability to extensively self-renew in vitro, while retaining multipotency, qualifies mesoangioblasts as a novel class of stem cells. Phenotype, properties and possible origin of mesoangioblasts are addressed in the first part of this paper. In the second part we will focus on the cell therapy approach for the treatment of Muscular Dystrophy and we will describe why mesangioblasts appear to be promising candidates for this strategy.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/tendencias , Células Madre Mesenquimatosas/fisiología , Enfermedades Musculares/terapia , Regeneración/fisiología , Animales , Biomarcadores/metabolismo , Vasos Sanguíneos/citología , Vasos Sanguíneos/embriología , Vasos Sanguíneos/metabolismo , Diferenciación Celular/fisiología , Vectores Genéticos/fisiología , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Enfermedades Musculares/fisiopatología , Sarcoglicanos/genética , Transfección/métodos , Transfección/tendencias
18.
Am J Physiol Cell Physiol ; 281(2): C690-9, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11443068

RESUMEN

Deficiency of delta-sarcoglycan (delta-SG), a component of the dystrophin-glycoprotein complex, causes cardiomyopathy and skeletal muscle dystrophy in Bio14.6 hamsters. Using cultured myotubes prepared from skeletal muscle of normal and Bio14.6 hamsters (J2N-k strain), we investigated the possibility that the delta-SG deficiency may lead to alterations in ionic conductances, which may ultimately lead to myocyte damage. In cell-attached patches (with Ba(2+) as the charge carrier), an approximately 20-pS channel was observed in both control and Bio14.6 myotubes. This channel is also permeable to K(+) and Na(+) but not to Cl(-). Channel activity was increased by pressure-induced stretch and was reduced by GdCl(3) (>5 microM). The basal open probability of this channel was fourfold higher in Bio14.6 myotubes, with longer open and shorter closed times. This was mimicked by depolymerization of the actin cytoskeleton. In intact Bio14.6 myotubes, the unidirectional basal Ca(2+) influx was enhanced compared with control. This Ca(2+) influx was sensitive to GdCl(3), signifying that stretch-activated cation channels may have been responsible for Ca(2+) influx in Bio14.6 hamster myotubes. These results suggest a possible mechanism by which cell damage might occur in this animal model of muscular dystrophy.


Asunto(s)
Cationes/metabolismo , Proteínas del Citoesqueleto/deficiencia , Canales Iónicos/metabolismo , Glicoproteínas de Membrana/deficiencia , Músculo Esquelético/metabolismo , Actinas/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Cricetinae , Citocalasina D/farmacología , Conductividad Eléctrica , Electrofisiología , Gadolinio/farmacología , Homeostasis , Membranas Intracelulares/metabolismo , Canales Iónicos/efectos de los fármacos , Canales Iónicos/fisiología , Cinética , Masculino , Técnicas de Placa-Clamp , Estimulación Física , Polímeros/metabolismo , Valores de Referencia , Sarcoglicanos
19.
Pflugers Arch ; 442(2): 161-70, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11417209

RESUMEN

Sarcoglycans (SGs) are components of the dystrophin-glycoprotein complex, genetic defects in which cause skeletal muscle dystrophy and cardiomyopathy in humans and animals. To obtain insight into the roles of SGs, we characterized properties of myotubes prepared from cells of the rat L6 line or primary myoblast cultures of rat gastrocnemius muscle that were made SG-deficient by treatment with antisense oligodeoxynucleotides (AS-ODNs). Immunoblot and immunoprecipitation analyses revealed that dystrophin and its remaining associated proteins were tightly associated in these cells despite SG deficiency. 45Ca2+ influx into SG AS-ODN-treated L6 myotubes under resting conditions was significantly higher (1.7-fold at 6 min) than in controls, suggesting that Ca2+ influx is activated in these SG-deficient myotubes. When these cells were subjected to cyclic elongation of up to 20% for 1 h, a marked increase in creatine phosphokinase (CK) release into the medium was observed. Nifedipine, tranilast, FK506 and E64 or intracellular loading with 1,2-bis(2-aminophenoxy)ethane- N,N,N',N'-tetraacetic acid, tetrakis(acetoxymethyl)ester (BAPTA/AM) reduced the stretch-induced CK release; a raised extracellular [Ca2+] increased CK release. The stretch-induced damage to SG-deficient myotubes thus appears to be caused by alterations in cell Ca2+ homeostasis. A similar abnormality in Ca2+ handling has been reported for myoctes from mdx mice or dystrophin-deficient patients, in whom SGs are also greatly reduced or absent. Thus it is possible that SG deficiency may play a critical role in the pathology of dystrophin-deficient muscle.


Asunto(s)
Glicoproteínas de Membrana/deficiencia , Músculos/metabolismo , Músculos/patología , Animales , Calcio/farmacocinética , Línea Celular , Membrana Celular/metabolismo , Creatina Quinasa/metabolismo , Distrofina/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Músculos/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Estimulación Física , Ratas , Receptores Colinérgicos/metabolismo
20.
Lab Invest ; 77(5): 489-502, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9389792

RESUMEN

Current information regarding the molecular and biochemical mechanisms of myocardial hypertrophy, as obtained from isolated cardiomyocytes and/or healthy animals with aortic banding, does not permit dissection of the hierarchical relationship among different steps and triggers of the pathogenic process in vivo. The aim of the present study was to depict the temporal relationship among myocardial structural and functional characteristics, the embryonic gene program, and transforming growth factor (TGF) beta 1 expression in euthyroid hereditary hypertrophic cardiomyopathic hamsters (CMPH). This investigation was performed using Western and Northern blot and in situ hybridization techniques. The results show that in CMPH, the severity of the hemodynamic overload is not related to any modification in structural myocardial characteristics (cardiac mass, cardiomyocyte dimensions, total RNA, and protein content), whereas an early activation of the embryonic gene program occurs in not yet overloaded 90-day-old CMPH (left ventricular end diastolic pressure < 15 mm Hg). In these animals, a 30% to 90% decrease in the alpha myosin heavy chain (alpha MHC) relative content was found in ventricles, whereas beta MHC increased 5-fold. In addition, the alpha skeletal actin expression was enhanced 2-fold versus age-matched controls. No modifications were observed in myosin function evaluated by in vitro motility assay, whereas the administration of L-thyroxine (100 micrograms/kg intraperitoneally daily) to CMPH was able to reinduce the ventricular expression of the alpha MHC isoform (5-fold increase). Conversely, no changes were found in alpha cardiac actin and myosin light chain 2 (MLC2) expression. A close temporal relationship occurred in CMPH ventricles between the re-expression of the embryonic gene program and a 3-fold enhancement of the expression of TGF beta 1. These results indicate that the CMPH provides a useful model for investigating the expression of embryonic genes in hypertrophic ventricles in the absence of mechanical and hormonal stimuli, and that TGF beta 1 is involved in regulating in vivo the "embryonic step" of myocardial hypertrophy. Furthermore, the study offers new insights into the pathophysiologic mechanisms leading to heart failure.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Regulación del Desarrollo de la Expresión Génica , Actinas/biosíntesis , Actinas/genética , Actinas/fisiología , Animales , Cardiomiopatía Hipertrófica/embriología , Cardiomiopatía Hipertrófica/enzimología , Cricetinae , ADN/análisis , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/enzimología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Hemodinámica , Hibridación in Situ , Isoenzimas/análisis , Miocardio/enzimología , Miocardio/metabolismo , Cadenas Pesadas de Miosina/análisis , Miosinas/análisis , Miosinas/fisiología , Tamaño de los Órganos , ARN/análisis , Hormonas Tiroideas/sangre , Factor de Crecimiento Transformador beta/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA