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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Emerg Med ; 76: 105-110, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38056055

RESUMO

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.


Assuntos
Traumatismos Craniocerebrais , Humanos , Estudos Prospectivos , Traumatismos Craniocerebrais/complicações , Traumatismos Craniocerebrais/diagnóstico por imagem , Anticoagulantes/efeitos adversos , Fatores de Risco , Estudos Retrospectivos
2.
Differentiation ; 133: 25-39, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37451110

RESUMO

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.


Assuntos
Células-Tronco Adultas , Paralisia Cerebral , Contratura , Humanos , Criança , Pré-Escolar , Paralisia Cerebral/patologia , Reprodutibilidade dos Testes , Músculo Esquelético/patologia , Contratura/patologia
3.
Histochem Cell Biol ; 139(1): 35-46, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22936275

RESUMO

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.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/enzimologia , Miócitos Cardíacos/enzimologia , Proteína Quinase C-épsilon/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Fator de Transcrição GATA4/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Proteína Quinase C-épsilon/genética , Interferência de RNA , Ratos , Ratos Wistar , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção
4.
Nat Med ; 12(10): 1147-50, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16980968

RESUMO

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.


Assuntos
Inibidores Enzimáticos/farmacologia , Músculos/enzimologia , Músculos/patologia , Distrofia Muscular Animal/tratamento farmacológico , Animais , Distrofina/genética , Fibrose/patologia , Folistatina/metabolismo , Ácidos Hidroxâmicos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Músculos/efeitos dos fármacos , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Fenilbutiratos/farmacologia , Sarcoglicanas/metabolismo , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/enzimologia , Ácido Valproico/farmacologia
5.
Cell Death Differ ; 15(9): 1417-28, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18497758

RESUMO

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.


Assuntos
Ventrículos do Coração/citologia , Miócitos Cardíacos/citologia , Células-Tronco/citologia , Animais , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Diferenciação Celular , Proliferação de Células , Endotélio Vascular/citologia , Ventrículos do Coração/crescimento & desenvolvimento , Humanos , Camundongos , Miocárdio/citologia , Técnicas de Patch-Clamp , Ratos , Células-Tronco/metabolismo , Células-Tronco/fisiologia
6.
J Nutr Health Aging ; 23(8): 739-745, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31560032

RESUMO

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.


Assuntos
Autofagia/fisiologia , Dieta/métodos , Músculo Esquelético/fisiopatologia , Humanos
7.
Nat Commun ; 10(1): 492, 2019 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-30700722

RESUMO

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.


Assuntos
Biologia Computacional/métodos , Terapia Genética/métodos , Músculo Esquelético/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Vetores Genéticos/genética , Humanos , Masculino , Camundongos , Camundongos SCID , Mutação/genética , Regiões Promotoras Genéticas/genética
8.
Cell Death Differ ; 14(6): 1162-71, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17347663

RESUMO

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.


Assuntos
Diferenciação Celular , Polpa Dentária/citologia , Osteoblastos/citologia , Osteogênese , Adipócitos/citologia , Adipócitos/metabolismo , Adulto , Animais , Técnicas de Cultura de Células , Células Cultivadas , Polpa Dentária/metabolismo , Polpa Dentária/ultraestrutura , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Microscopia Eletrônica de Transmissão , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Osteoblastos/metabolismo , Osteoblastos/ultraestrutura , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Células-Tronco/metabolismo , Células-Tronco/ultraestrutura , Células Estromais/citologia , Células Estromais/metabolismo , Células Estromais/ultraestrutura , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Fatores de Tempo
9.
FASEB J ; 21(13): 3573-83, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17601985

RESUMO

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."


Assuntos
Inibidores Enzimáticos/farmacologia , Coração/efeitos dos fármacos , Miocárdio/citologia , Células-Tronco Pluripotentes/citologia , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Compostos de Vanádio/farmacologia , Animais , Sequência de Bases , Ciclo Celular , Linhagem Celular , Primers do DNA , Citometria de Fluxo , Imunofluorescência , Expressão Gênica , Camundongos , Miocárdio/metabolismo , Fenótipo , Células-Tronco Pluripotentes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Cell Transplant ; 16(6): 563-77, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17912948

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Distrofia Muscular de Duchenne/terapia , Mioblastos Esqueléticos/transplante , Peptídeos/metabolismo , Antígeno AC133 , Adolescente , Antígenos CD/classificação , Antígenos CD/isolamento & purificação , Criança , Método Duplo-Cego , Estudos de Viabilidade , Seguimentos , Glicoproteínas/classificação , Glicoproteínas/isolamento & purificação , Humanos , Separação Imunomagnética/classificação , Imunofenotipagem/classificação , Injeções Intramusculares , Masculino , Contração Muscular/fisiologia , Músculo Esquelético/citologia , Distrofia Muscular de Duchenne/patologia , Mioblastos Esqueléticos/citologia , Peptídeos/classificação , Peptídeos/isolamento & purificação , Transplante de Células-Tronco , Células-Tronco/citologia , Transplante Autólogo , Transplante Homólogo/efeitos adversos , Resultado do Tratamento
11.
Arch Ital Biol ; 143(3-4): 235-42, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16097501

RESUMO

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.


Assuntos
Transplante de Células-Tronco Mesenquimais/tendências , Células-Tronco Mesenquimais/fisiologia , Doenças Musculares/terapia , Regeneração/fisiologia , Animais , Biomarcadores/metabolismo , Vasos Sanguíneos/citologia , Vasos Sanguíneos/embriologia , Vasos Sanguíneos/metabolismo , Diferenciação Celular/fisiologia , Vetores Genéticos/fisiologia , Humanos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Doenças Musculares/fisiopatologia , Sarcoglicanas/genética , Transfecção/métodos , Transfecção/tendências
12.
Biotech Histochem ; 90(6): 424-31, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25893542

RESUMO

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.


Assuntos
Órgãos Bioartificiais , Microscopia Confocal/métodos , Músculo Esquelético/anatomia & histologia , Músculo Esquelético/metabolismo , Animais , Células Cultivadas , Proteínas de Fluorescência Verde/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Imageamento Tridimensional , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Proteínas Recombinantes/metabolismo , Engenharia Tecidual
13.
Endocrinology ; 137(10): 4339-50, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8828494

RESUMO

Atrial natriuretic factor (ANF) and ANF receptor C (ANF.RC) expression have been investigated in healthy and cardiomyopathic hamsters (CMPH) with widespread necrosis of the diaphragm and myocardium leading to respiratory and heart failure. ANF- and ANF.RC-producing cells were localized in different structures of the respiratory system, and the regulation of their expression by the individual and/or combined action of hypoxia and hemodynamic overload was analyzed. The study was performed in 20-, 90-, and 150-day-old animals using immunohistochemistry, in situ hybridization, Northern blot, and RIA analyses. ANF was shown to be expressed in the tracheo-bronchial epithelium and muscle and, to a lesser extent, in the alveolar wall and muscular media of the pulmonary arteries and extraparenchymal pulmonary veins in both healthy hamsters and CMPH. In 150-day-old CMPH, hypoxia (PaO2 < 50 mm Hg) induced a 10-fold increase in ANF messenger RNA accumulation and a 6-fold increase in the immunoreactive ANF (IR-ANF) concentration in lungs, as quantitated by RIA. As plasma IR-ANF concentrations were elevated in all CMPH age groups, it was most likely produced by the myocardium. ANF.RC messenger RNA was homogeneously distributed throughout the entire respiratory system and was increased 2-fold in hypoxic 150-day-old CMPH only. These results suggest that ANF originating in the respiratory system exerts only paracrine effects on different structures of the respiratory system in addition to the action of circulating ANF. Hemodynamic overload (left ventricular end-diastolic pressure, 17.20 +/- 3.80 mm Hg) might contribute to enhanced ANF gene expression only in extraparenchymal pulmonary vein walls of 150-day-old CMPH. We also propose that ANF.RC overexpression might be a protective mechanism operated via either ANF clearance or inhibition of adenylate cyclase activity to counteract exaggerated smooth muscle relaxation.


Assuntos
Fator Natriurético Atrial/genética , Cardiomiopatias/metabolismo , Expressão Gênica , Hipóxia/metabolismo , Receptores do Fator Natriurético Atrial/genética , Sistema Respiratório/metabolismo , Animais , Fator Natriurético Atrial/metabolismo , Cardiomiopatias/sangue , Cardiomiopatias/fisiopatologia , Cricetinae , Gases/sangue , Hemodinâmica , Concentração de Íons de Hidrogênio , Imuno-Histoquímica , Hibridização In Situ , Mesocricetus , RNA Mensageiro/metabolismo , Receptores do Fator Natriurético Atrial/metabolismo , Distribuição Tecidual
14.
Cell Death Dis ; 5: e1448, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25299773

RESUMO

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.


Assuntos
Desenvolvimento Muscular , Receptor Notch1/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Animais , Proteínas de Ligação ao Cálcio , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Receptor Notch1/genética , Células-Tronco/metabolismo
15.
Cell Transplant ; 21(9): 1945-67, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22513051

RESUMO

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.


Assuntos
Distrofia Muscular Animal/patologia , Miocárdio/citologia , Miocárdio/patologia , Células-Tronco/patologia , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Modelos Animais de Doenças , Cães , Feminino , Citometria de Fluxo , Humanos , Masculino , Camundongos , Camundongos SCID , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Miocárdio/metabolismo , Ratos , Células-Tronco/metabolismo , Transcriptoma
16.
Res Vet Sci ; 91(1): 18-24, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20732703

RESUMO

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.


Assuntos
Tecido Adiposo/citologia , Criopreservação/veterinária , Células-Tronco Mesenquimais/fisiologia , Fosfatase Alcalina/metabolismo , Animais , Antígenos de Superfície/metabolismo , Técnicas de Cultura de Células/veterinária , Diferenciação Celular , Cães , Feminino , Citometria de Fluxo/veterinária , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Reação em Cadeia da Polimerase/veterinária , Análise de Sequência de RNA/veterinária , Telomerase/metabolismo
17.
Cell Death Differ ; 17(8): 1222-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19876070

RESUMO

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.


Assuntos
Distrofias Musculares/terapia , Transplante de Células-Tronco , Animais , Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/transplante , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia
19.
J Biomed Mater Res A ; 84(4): 1094-101, 2008 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17685407

RESUMO

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.


Assuntos
Materiais Biocompatíveis/química , Desenvolvimento Muscular , Poliésteres/química , Poliuretanos/química , Animais , Adesão Celular , Linhagem Celular , Desenho de Equipamento , Teste de Materiais , Camundongos , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Ratos , Engenharia Tecidual/métodos
20.
Pflugers Arch ; 442(2): 161-70, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11417209

RESUMO

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.


Assuntos
Glicoproteínas de Membrana/deficiência , Músculos/metabolismo , Músculos/patologia , Animais , Cálcio/farmacocinética , Linhagem Celular , Membrana Celular/metabolismo , Creatina Quinase/metabolismo , Distrofina/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Músculos/efeitos dos fármacos , Oligonucleotídeos Antissenso/farmacologia , Estimulação Física , Ratos , Receptores Colinérgicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA