Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-31871077

RESUMEN

We analyzed the use of isavuconazole (ISA) as treatment or prophylaxis for invasive fungal disease (IFD) in children with hemato-oncologic diseases. A multicentric retrospective analysis was performed among centers belonging to the Italian Association for Pediatric Hematology and Oncology (AIEOP). Pharmacokinetic (PK) monitoring was applied by a high-performance liquid chromatography-tandem mass spectrometry (HLPC-MS/MS) assay. Twenty-nine patients were studied: 10 during chemotherapy and 19 after allogeneic hematopoietic stem cell transplantation (HSCT). The patients consisted of 20 males and 9 females with a median age of 14.5 years (age range, 3 to 18 years) and a median body weight of 47 kg (body weight range, 15 to 80 kg). ISA was used as prophylaxis in 5 patients and as treatment in 24 cases (20 after therapeutic failure, 4 as first-line therapy). According to European Organization for Research and Treatment of Cancer (EORTC) criteria, we registered 5 patients with proven IFD, 9 patients with probable IFD, and 10 patients with possible IFD. Patients with a body weight of <30 kg received half the ISA dose; the others received ISA on the adult schedule (a 200-mg loading dose every 8 h on days 1 and 2 and a 200-mg/day maintenance dose); for all but 10 patients, the route of administration switched from the intravenous route to the oral route during treatment. ISA was administered for a median of 75.5 days (range, 6 to 523 days). The overall response rate was 70.8%; 12 patients with IFD achieved complete remission, 5 achieved partial remission, 5 achieved progression, and 3 achieved stable IFD. No breakthrough infections were registered. PK monitoring of 17 patients revealed a median ISA steady-state trough concentration of 4.91 mg/liter (range, 2.15 to 8.54 mg/liter) and a concentration/dose (in kilograms) ratio of 1.13 (range, 0.47 to 3.42). Determination of the 12-h PK profile was performed in 6 cases. The median area under the concentration-time curve from 0 to 12 h was 153.16 mg·h/liter (range, 86.31 to 169.45 mg·h/liter). Common Terminology Criteria for Adverse Events grade 1 to 3 toxicity (increased transaminase and/or creatinine levels) was observed in 6 patients, with no drug-drug interactions being seen in patients receiving immunosuppressants. Isavuconazole may be useful and safe in children with hemato-oncologic diseases, even in the HSCT setting. Prospective studies are warranted.


Asunto(s)
Antifúngicos/farmacocinética , Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas , Infecciones Fúngicas Invasoras/tratamiento farmacológico , Nitrilos/farmacocinética , Piridinas/farmacocinética , Triazoles/farmacocinética , Administración Intravenosa , Administración Oral , Adolescente , Antifúngicos/sangre , Antifúngicos/farmacología , Aspergillus/efectos de los fármacos , Aspergillus/crecimiento & desarrollo , Niño , Preescolar , Esquema de Medicación , Femenino , Neoplasias Hematológicas/microbiología , Neoplasias Hematológicas/patología , Humanos , Infecciones Fúngicas Invasoras/microbiología , Infecciones Fúngicas Invasoras/patología , Masculino , Pruebas de Sensibilidad Microbiana , Mucor/efectos de los fármacos , Mucor/crecimiento & desarrollo , Nitrilos/sangre , Nitrilos/farmacología , Penicillium/efectos de los fármacos , Penicillium/crecimiento & desarrollo , Piridinas/sangre , Piridinas/farmacología , Estudios Retrospectivos , Trasplante Homólogo , Triazoles/sangre , Triazoles/farmacología
2.
Am J Nephrol ; 33(3): 239-49, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21358177

RESUMEN

BACKGROUND: Ischemia-reperfusion (I/R) is present at various degrees in kidney transplants. I/R plays a major role in early function and long-term survival of renal allograft. The purpose of our study was to determine if immunosuppressants modulate I/R in a model that separates I/R from all immune responses. METHODS: Sprague-Dawley rats with monolateral renal I/R received daily cyclosporine (A), tacrolimus (B), sirolimus (C) or saline (D). Sham-operated rats received saline (E). After 30 days, glomerular filtration rate for each kidney was measured by inulin clearance. Kidney injury was examined, and TGF-ß, fibronectin and metalloproteases were evaluated by real-time PCR, Western blot and zymography. RESULTS: Sirolimus, but not cyclosporine and tacrolimus, prevented a glomerular filtration rate decrease in I/R kidneys (403 ± 303 vs. 1,006 ± 484 µl/min, p < 0.05; 126 ± 170 vs. 567 ± 374 µl/min, p < 0.05; 633 ± 293 vs. 786 ± 255; A, B and C group, respectively, I/R vs. contralateral kidneys). Sirolimus reduced ED-1+ cell infiltrate, interstitial fibrosis and intimal thickening of small vessels observed in I/R kidneys of controls and calcineurin inhibitor-treated rats. Tacrolimus and cyclosporine increased fibronectin and TGF-ß expression and matrix deposition. Only sirolimus increased metalloprotease activity. CONCLUSIONS: Sirolimus but not calcineurin inhibitors prevented I/R-induced kidney injury.


Asunto(s)
Ciclosporina/uso terapéutico , Inmunosupresores/uso terapéutico , Enfermedades Renales/prevención & control , Daño por Reperfusión/prevención & control , Sirolimus/uso terapéutico , Tacrolimus/uso terapéutico , Animales , Tasa de Filtración Glomerular , Riñón/patología , Enfermedades Renales/patología , Pruebas de Función Renal , Masculino , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/patología
3.
Cell Death Dis ; 1: e45, 2010 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21364651

RESUMEN

Protein kinase Cs (PKCs) constitute a family of serine/threonine kinases, which has distinguished and specific roles in regulating cardiac responses, including those associated with heart failure. We found that the PKCθ isoform is expressed at considerable levels in the cardiac muscle in mouse, and that it is rapidly activated after pressure overload. To investigate the role of PKCθ in cardiac remodeling, we used PKCθ(-/-) mice. In vivo analyses of PKCθ(-/-) hearts showed that the lack of PKCθ expression leads to left ventricular dilation and reduced function. Histological analyses showed a reduction in the number of cardiomyocytes, combined with hypertrophy of the remaining cardiomyocytes, cardiac fibrosis, myofibroblast hyper-proliferation and matrix deposition. We also observed p38 and JunK activation, known to promote cell death in response to stress, combined with upregulation of the fetal pattern of gene expression, considered to be a feature of the hemodynamically or metabolically stressed heart. In keeping with these observations, cultured PKCθ(-/-) cardiomyocytes were less viable than wild-type cardiomyocytes, and, unlike wild-type cardiomyocytes, underwent programmed cell death upon stimulation with α1-adrenergic agonists and hypoxia. Taken together, these results show that PKCθ maintains the correct structure and function of the heart by preventing cardiomyocyte cell death in response to work demand and to neuro-hormonal signals, to which heart cells are continuously exposed.


Asunto(s)
Isoenzimas/metabolismo , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Proteína Quinasa C/metabolismo , Remodelación Ventricular/fisiología , Animales , Cardiomegalia/complicaciones , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/enzimología , Cardiomegalia/fisiopatología , Recuento de Células , Supervivencia Celular , Activación Enzimática , Fibroblastos/enzimología , Fibroblastos/patología , Eliminación de Gen , Hemodinámica , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Miocardio/enzimología , Miocardio/patología , Presión , Proteína Quinasa C-theta , Ultrasonografía , Disfunción Ventricular Izquierda/complicaciones , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/fisiopatología
4.
Eur J Histochem ; 51 Suppl 1: 35-43, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17703592

RESUMEN

One of the most exciting aspirations of current medical science is the regeneration of damaged body parts. The capacity of adult tissues to regenerate in response to injury stimuli represents an important homeostatic process that until recently was thought to be limited in mammals to tissues with high turnover such as blood and skin. However, it is now generally accepted that each tissue type, even those considered post-mitotic, such as nerve or muscle, contains a reserve of undifferentiated progenitor cells, loosely termed stem cells, participating in tissue regeneration and repair. Skeletal muscle regeneration is a coordinate process in which several factors are sequentially activated to maintain and preserve muscle structure and function upon injury stimuli. In this review, we will discuss the role of stem cells in muscle regeneration and repair and the critical role of specific factors, such as IGF-1, vasopressin and TNF-alpha, in the modulation of the myogenic program and in the regulation of muscle regeneration and homeostasis.


Asunto(s)
Envejecimiento/fisiología , Músculo Esquelético/fisiología , Enfermedades Neuromusculares/fisiopatología , Regeneración , Animales , Diferenciación Celular , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Madre/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Vasopresinas/metabolismo
5.
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
6.
Transplant Proc ; 38(6): 1809-11, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16908288

RESUMEN

Malononitrilamide 715 (FK778) is a new class of low molecular weight immunosuppressant. Experimental studies in heart, liver, and kidney transplantation have shown a strong synergism when FK778 is used in combination with tacrolimus and when its administration is delayed by 7 days after the transplant. Following this indication, in a swine model of orthotopic small bowel transplantation (SBT), we assessed the efficacy of combined low dose tacrolimus and FK778 administered from day 0 or day 7. The entire small bowel was replaced in 16 piglets: group 1 (n = 5), no immunosuppression; group 2 (n = 6) oral tacrolimus to maintain whole blood trough levels between 5 and 15 ng/mL plus FK778 4 mg/kg per day; group 3 (n = 6) oral tacrolimus as in group 2 plus FK778 4 mg/kg per day administered after a 7-day delay posttransplant. The median survival was 8 days in group 1, 60 days in group 2, and 13 days in group 3. The differences between group 2 and 1 and between group 2 and 3 are statistically significant. Three episodes of major bacterial infection were detected in both group 2 and 3 (0.5 episode/animal). The infectious-related mortality was 0% in group 2 and 50% in group 3 (P < .05). Acute cellular rejection was absent or mild in all group 2 and 3 stomal biopsies. In conclusion, combining tacrolimus and FK778 allowed prolonged survival after SBT in swine when FK778 was started at the time of SBT. The delayed administration of FK778 resulted in a high incidence of lethal infectious complications.


Asunto(s)
Supervivencia de Injerto/inmunología , Intestino Delgado/trasplante , Isoxazoles/uso terapéutico , Tacrolimus/uso terapéutico , Alquinos , Animales , Quimioterapia Combinada , Rechazo de Injerto/inmunología , Supervivencia de Injerto/efectos de los fármacos , Modelos Animales , Nitrilos , Análisis de Supervivencia , Porcinos , Inhibidores de Tripsina/uso terapéutico
7.
Transplant Proc ; 38(6): 1812-4, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16908289

RESUMEN

The intestine is a highly immunogenic organ that requires heavy immunosuppression (IS); therefore corticosteroid withdrawal after clinical small bowel transplantation (SBT) has not been standardized. In this study, we compared different immunosuppressive regimens (none with steroid or induction treatment) in a SBT pig model. Large White unrelated piglets were transplanted and divided into four groups as follow: group 1 (n = 3): no IS; group 2 (n = 10): IS with tacrolimus only; group 3 (n = 10): IS with tacrolimus and mycophenolate mofetil; group 4 (n = 5): IS with tacrolimus and rapamycin. Follow-up time was 30 days. All IS drugs were given orally; tacrolimus whole blood levels ranged between 5 and 15 ng/mL in all groups except for group 2 whose tacrolimus whole blood levels ranged between 15 and 25 ng/mL. Group 1 pigs died of graft acute rejection (ACR) after a median of 12 days. Overall survival in groups 2, 3, and 4 at day 30 was 40%, 80%, and 60%, respectively. Biochemical parameters, including glycemia and cholesterol, were into the normal range with no significant differences between groups. At the end of the study, one animal in group 2 and another one in group 4 showed histological signs of moderate to severe ACR. The incidence of infection was higher in group 2 (2.1 episodes/pig) compared to group 3 (1.25) and group 4 (1.6). This large-animal study demonstrates that tacrolimus-based IS without corticosteroids allows, in the early postoperative period (30 days) after SBT, good survival rates without an increased risk in the incidence of rejection.


Asunto(s)
Supervivencia de Injerto/inmunología , Inmunosupresores/uso terapéutico , Intestino Delgado/trasplante , Corticoesteroides , Animales , Supervivencia de Injerto/efectos de los fármacos , Modelos Animales , Porcinos , Trasplante Homólogo/inmunología , Resultado del Tratamiento
8.
J Cell Physiol ; 207(2): 379-88, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16419034

RESUMEN

Adult skeletal muscle fibers can be divided into fast and slow twitch subtypes on the basis of specific contractile and metabolic properties, and on distinctive patterns of muscle gene expression. The calcium, calmodulin-dependent protein phosphatase, calcineurin, stimulates slow fiber-specific genes (myoglobin (Mb), troponin I slow) in cultured skeletal muscle cells, as well as in transgenic mice, through the co-operation of peroxisome-proliferation-activator receptor gamma co-activator 1alpha (PGC1alpha) myocyte enhancer factor 2 (MEF2), and nuclear factor of activated T cells (NFAT) transcription factors. Specific protein kinase C isoforms have been shown to functionally co-operate with calcineurin in different cellular models. We investigated whether specific protein kinase C isoforms are involved in calcineurin-induced slow skeletal muscle gene expression. By pharmacological inhibition or exogenous expression of mutant forms, we show that protein kinase C theta (the protein kinase C isoform predominantly expressed in skeletal muscle) is required and co-operates with calcineurin in the activation of the Mb promoter, as well as in the induction of slow isoforms of myosin and troponin I expression, in cultured muscle cells. This co-operation acts primarily regulating MEF2 activity, as shown by using reporter gene expression driven by the Mb promoter mutated in the specific binding sites. MEF2 activity on the Mb promoter is known to be dependent on both PGC1alpha and inactivation of histone deacetylases (HDACs) activity. We show in this study that protein kinase C theta is required for, even though it does not co-operate in, PGC1alpha-dependent Mb activation. Importantly, protein kinase C theta regulates the HDAC5 nucleus/cytoplasm location. We conclude that protein kinase C theta ensures maximal activation of MEF2, by regulating both MEF2 transcriptional complex formation and HDACs nuclear export.


Asunto(s)
Calcineurina/farmacología , Isoenzimas/metabolismo , Fibras Musculares de Contracción Lenta/metabolismo , Mioblastos/metabolismo , Proteína Quinasa C/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Calcineurina/genética , Carbazoles/farmacología , Línea Celular , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Indoles/farmacología , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Factores de Transcripción MEF2 , Ratones , Fibras Musculares de Contracción Lenta/efectos de los fármacos , Mutación , Mioblastos/citología , Mioblastos/efectos de los fármacos , Factores Reguladores Miogénicos/genética , Mioglobina/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/genética , Proteína Quinasa C-theta , Proteínas de Unión al ARN/genética , Ratas , Transactivadores/genética , Factores de Transcripción/genética , Transfección , Troponina I/genética
9.
Transplant Proc ; 37(6): 2719-21, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16182793

RESUMEN

In a swine model of orthotopic small bowel transplantation, we assessed the efficacy of combined immunosuppressive therapy with low-dose tacrolimus plus FK778, compared with high-dose tacrolimus monotherapy. The small bowel was replaced in 23 piglets: group 1 (n = 5), no immunosuppression; group 2 (n = 12), oral tacrolimus to maintain whole blood trough levels between 15 and 25 ng/mL; and group 3 (n = 6), oral FK778 4 mg/kg/d, plus oral tacrolimus to maintain whole blood trough levels between 5 and 15 ng/mL. Follow-up time was limited to 60 days. Overall survival rates at 30 and 60 days were 0% and 0% in group 1, 30% and 0% in group 2, and 66% and 66% in group 3, respectively. The median survival time was 11 days in group 1, 28 days in group 2, and more than 60 days in group 3. The differences between groups 3 and 1 and between groups 3 and 2 were statistically significant. The numbers of major bacterial infections were 19 in group 2 (1.9 episodes per animal) and 3 in group 3 (0.75 episodes per animal). The infectious-related mortality rate was 70% in group 2 (7 cases) and 0% in group 3 (P < .05). Acute cellular rejection was absent or mild in 85% of group 2 stomal biopsy specimens and in 100% of group 3 biopsy specimens. In conclusion, combination therapy of low-dose tacrolimus is potentiated by FK778 to prevent acute cellular rejection and prolong small bowel transplant survival in pigs.


Asunto(s)
Inmunosupresores/uso terapéutico , Intestino Delgado/trasplante , Isoxazoles/uso terapéutico , Alquinos , Animales , Modelos Animales , Nitrilos , Análisis de Supervivencia , Porcinos , Trasplante Homólogo/inmunología , Trasplante Homólogo/mortalidad
10.
Transplant Proc ; 37(6): 2722-7, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16182794

RESUMEN

FK778 is a new immunosuppressive agent, derived from the leflunomide-active metabolite A77 1726. It inhibits de novo pyrimidine nucleotide synthesis showing efficacy in the prevention and treatment of rejection in experimental transplant models. The aim of this work was to develop an HPLC-MS method to measure FK778 in plasma for pharmacokinetic studies. The equipment used for mass evaluation was an HLPC coupled to an ion trap analyzer through an electrospray source. After precipitation of plasma proteins with acetonitrile, the supernatant was injected onto an analytical RP-C18 column. Chromatographic separation was performed under isocratic conditions, using a mobile phase consisting of ammonium acetate buffer and acetonitrile (55:45. vol/vol). MS detection was performed in the negative ionization mode by monitoring the molecular ion of FK778 (m/z 307) and IS (m/z 269), using selected ion monitoring for both. However, we observed peaks corresponding to dimers, trimers, and tetramers of FK778 (m/z 637, m/z 945, m/z 1274). The HPLC-MS method was applied to pharmacokinetics in animal models showing comparable results to those obtained by an HPLC-UV assay at 290 nm. Good agreement was observed in the plasma FK778 concentration versus time curves. The rapid preparation of samples and the short run-time make this method attractive for use in clinical practice.


Asunto(s)
Trasplante de Corazón/inmunología , Inmunosupresores/sangre , Isoxazoles/sangre , Trasplante de Riñón/inmunología , Alquinos , Calibración , Cromatografía Líquida de Alta Presión , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/inmunología , Humanos , Leflunamida , Espectrometría de Masas , Nitrilos , Espectrometría de Masa por Ionización de Electrospray
11.
Transplant Proc ; 37(4): 1697-9, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15919435

RESUMEN

To assess the efficacy and safety of a primary immunosuppressive regimen with tacrolimus (Tac) and low-dose mycophenolate mofetil (MMF) without steroids and to determine the exposure to mycophenolic acid (MPA) in the early postoperative period, we performed a single-center, randomized 1:1, open-label, controlled study planned to be 60 liver transplantation patients randomized into 2 groups: group A, tacrolimus + MMF (750 mg orally twice a day); and group B, tacrolimus + MMF (750 mg orally twice a day) + steroids. After an interim analysis by the ethical committee patient enrollment was stopped. Data from 30 patients (12 in group A and 18 in group B with a mean follow-up period of 31 +/- 7 months) showed a patient survival rate of 91.7% in group A and 100% in group B and a graft survival rate of 91.7% and 88.9%, respectively. Nine patients (75%) in group A suffered an acute rejection episode, whereas in group B only 3 patients (16.7%) showed acute rejection (P = .002). All rejection episodes occurred in both groups at 1 week after transplantation. The difference in histological grading was statistically significant (P = .021). The toxicity profiles were similar in both groups. A primary immunosuppressive regimen based on Tac and low-dose MMF without steroids is safe but unable to prevent acute rejection at 1 week after transplantation even if early acute rejection does not affect the outcome in terms of morbidity and graft or patient survival.


Asunto(s)
Rechazo de Injerto/prevención & control , Trasplante de Hígado/inmunología , Ácido Micofenólico/análogos & derivados , Tacrolimus/uso terapéutico , Enfermedad Aguda , Corticoesteroides/efectos adversos , Adulto , Área Bajo la Curva , Quimioterapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Inmunosupresores/uso terapéutico , Trasplante de Hígado/mortalidad , Masculino , Persona de Mediana Edad , Ácido Micofenólico/sangre , Ácido Micofenólico/farmacocinética , Ácido Micofenólico/uso terapéutico , Periodo Posoperatorio , Análisis de Supervivencia
12.
J Cell Physiol ; 202(3): 787-95, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15389566

RESUMEN

Calcium plays a pivotal role in the establishment of the differentiated phenotype in myogenic cells but the involved molecular mechanisms are still matter of debate. Here we studied the effects of exposing L6-C5 myogenic cells to high extracellular Ca2+ concentration ([Ca2+]o), which induces an increase of intracellular calcium ([Ca2+]i) without involving Ca2+ release from the intracellular stores but exclusively due to plasma membrane influx (Naro et al., 2003). Exposure of L6-C5 cells to [Ca2+]o up to 20 mM for 30 min, before shifting them into a differentiative medium, induced the appearance of multinucleated, myosin-positive myotubes, much larger than in control cells with an increased protein/DNA ratio. These large myotubes showed nuclear accumulation of the hypertrophy marker GATA-2. The hypertrophic growth of these cells was blocked by cyclosporin A (CsA), FK506, or overexpression of a calcineurin-dominant negative protein, suggesting the involvement in this process of the Ca2+ responsive phosphatase calcineurin. Furthermore, transient exposure of L6-C5 cells to high [Ca2+]o increased the expression of luciferase reporter driven by myoglobin (Mb) and beta-MHC promoters but not IIB-MHC and MCK promoters. Luciferase transcription driven by CK promoter was, instead, enhanced by mobilizing Ca2+ from the intracellular stores. These data indicate that a transient increase of [Ca2+]i due to plasma-membrane influx is sufficient to induce a hypertrophic phenotype and an increased expression of slow-fiber genes but not fast-fiber genes.


Asunto(s)
Calcio/metabolismo , Regulación de la Expresión Génica , Fibras Musculares Esqueléticas/citología , Transcripción Genética , Animales , Calcineurina/metabolismo , Diferenciación Celular/fisiología , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factor de Transcripción GATA2 , Hipertrofia , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares de Contracción Lenta/fisiología , Miosinas/genética , Miosinas/metabolismo , Regiones Promotoras Genéticas , Ratas , Transducción de Señal/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
13.
J Cell Physiol ; 196(1): 89-97, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12767044

RESUMEN

Protein kinase C theta (PKC-theta) is the PKC isoform predominantly expressed in skeletal muscle, and it is supposed to mediate many signals necessary for muscle histogenesis and homeostasis, such as TGFbeta, nerve-dependent signals and insulin. To study the role of PKC-theta in these mechanisms we generated transgenic mice expressing a "kinase dead" mutant form of PKC-theta (PKC-thetaK/R), working as "dominant negative," specifically in skeletal muscle. These mice are viable and fertile, however, by the 6-7 months of age, they gain weight, mainly due to visceral fat deposition. Before the onset of obesity (4 months of age), they already show increased fasting and fed insulin levels and reduced insulin-sensitivity, as measured by ipITT, but normal glucose tolerance, as measured by ipGTT. After the 6-7 months of age, transgenic mice develop hyperinsulinemia in the fasting and fed state. The ipGTT revealed in the transgenic mice both hyperglycemia and hyperinsulinemia. At the molecular level, impaired activation of the IR/IRS/PI3K pathway and a significant decrease both in the levels and in insulin-stimulated activation of the serine/threonine kinase Akt were observed. Taken together these data demonstrate that over-expression of dominant negative PKC-theta in skeletal muscle causes obesity associated to insulin resistance, as demonstrated by defective receptor and post-receptorial activation of signaling cascade.


Asunto(s)
Modelos Animales de Enfermedad , Genes Dominantes/genética , Resistencia a la Insulina , Isoenzimas/genética , Isoenzimas/metabolismo , Músculo Esquelético/enzimología , Obesidad , Proteína Quinasa C/genética , Proteína Quinasa C/metabolismo , Animales , Glucosa/metabolismo , Insulina/farmacología , Resistencia a la Insulina/genética , Ratones , Ratones Transgénicos , Mutación , Obesidad/genética , Fenotipo , Proteína Quinasa C-theta , Transducción de Señal/efectos de los fármacos
14.
Toxicol Appl Pharmacol ; 175(3): 226-33, 2001 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-11559021

RESUMEN

Polychlorinated biphenyls (PCBs) are ubiquitous and persistent pollutants whose role in developmental toxicity is of great concern. The observation that the offspring of PCB-exposed mothers (both in humans and rodents) display reduced body mass prompted us to investigate the effects of commercial mixtures of PCB congeners (Aroclor 1232, 1254, and 1262) on differentiation of both a myogenic cell line and primary myogenic cell cultures. The fusion of L6 myoblasts into multinucleated myotubes and the increase of creatine kinase (CK) activity were dose-dependently inhibited by Aroclor 1254 at concentrations (0.1-4 microg/ml) that caused no effect on cell density. Ultrastructural analysis demonstrated that Aroclor 1254 also prevented the accumulation of contractile filaments while inducing hypertrophy of the smooth endoplasmic reticulum and appearance of membrane-filled autophagosomes. Half-maximal inhibition (IC50) of CK activity accumulation occurred at 0.01 microg/ml for Aroclor 1262, 2 microg/ml for Aroclor 1254, and 8 microg/ml for Aroclor 1232. Aroclor-dependent inhibition of myogenic differentiation was also shown by the reduced expression and nuclear accumulation of beta-galactosidase in primary cultures of fetal myoblasts from transgenic mice expressing this reporter gene under the control of the myosin light chain promoter. These data show that skeletal muscle differentiation is specifically impaired by PCBs and may explain the reported depression of body mass growth in PCB-exposed offspring at birth. Furthermore, myogenic cell cultures are highly sensitive to PCBs and allow the detection of biological effects of environmental levels of these pollutants.


Asunto(s)
Arocloros/toxicidad , Diferenciación Celular/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Músculo Esquelético/efectos de los fármacos , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/ultraestructura , Animales , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/enzimología , Creatina Quinasa/antagonistas & inhibidores , Creatina Quinasa/metabolismo , ADN/biosíntesis , Relación Dosis-Respuesta a Droga , Retículo Endoplásmico Liso/efectos de los fármacos , Retículo Endoplásmico Liso/ultraestructura , Insulina/farmacología , Ratones , Ratones Transgénicos , Músculo Esquelético/citología , Músculo Esquelético/enzimología , Fagosomas/efectos de los fármacos , Fagosomas/ultraestructura , Ratas , Vasopresinas/farmacología , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
16.
Nat Genet ; 27(2): 195-200, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11175789

RESUMEN

Aging skeletal muscles suffer a steady decline in mass and functional performance, and compromised muscle integrity as fibrotic invasions replace contractile tissue, accompanied by a characteristic loss in the fastest, most powerful muscle fibers. The same programmed deficits in muscle structure and function are found in numerous neurodegenerative syndromes and disease-related cachexia. We have generated a model of persistent, functional myocyte hypertrophy using a tissue-restricted transgene encoding a locally acting isoform of insulin-like growth factor-1 that is expressed in skeletal muscle (mIgf-1). Transgenic embryos developed normally, and postnatal increases in muscle mass and strength were not accompanied by the additional pathological changes seen in other Igf-1 transgenic models. Expression of GATA-2, a transcription factor normally undetected in skeletal muscle, marked hypertrophic myocytes that escaped age-related muscle atrophy and retained the proliferative response to muscle injury characteristic of younger animals. The preservation of muscle architecture and age-independent regenerative capacity through localized mIgf-1 transgene expression suggests clinical strategies for the treatment of age or disease-related muscle frailty.


Asunto(s)
Senescencia Celular , Factor I del Crecimiento Similar a la Insulina/genética , Músculo Esquelético/fisiología , Regeneración , Animales , Caquexia , Proteínas de Unión al ADN/genética , Factor de Transcripción GATA2 , Hipertrofia , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas , Tamaño de los Órganos , Isoformas de Proteínas , Distribución Tisular , Factores de Transcripción/genética
17.
Ther Drug Monit ; 22(6): 712-5, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11128239

RESUMEN

The authors performed a comparative analysis of 60 whole blood samples containing cyclosporine (CsA) from heart transplant (HTx) recipients (n = 60) by the two "specific" monoclonal immunoassays, enzyme-multiplied immunoassay technique (EMIT) and fluorescence polarization immunoassay (S-FPIA), using the Altman-Bland approach based on graphical techniques and simple calculations. The CsA blood concentrations measured by S-FPIA [mean (SD): 268.1 (108.8) ng/mL] showed a statistically significant difference (P < 0.001) from the corresponding concentrations measured by EMIT [219.6 (118.7) ng/mL]. The CsA concentrations were 27% (median) higher when determined by monoclonal S-FPIA than by EMIT. The comparison between EMIT and S-FPIA showed a good correlation (S-FPIA conc. (ng/mL) = EMIT conc. (ng/mL) x 0.88 + 76.1, r = 0.96, P < 0.001). However, a high correlation does not mean that the two methods agree, and their use as interchangeable might be misleading. The authors summarized the degree of agreement by calculating the bias estimated by the mean difference (d) and the standard deviation of the difference (SD). For CsA concentration data, the mean difference (S-FPIA minus EMIT) is +49.9 ng/mL and SD is 31.2 ng/mL. Altman-Bland analysis indicates considerable lack of agreement between EMIT and S-FPIA, with discrepancies of more than 100 ng/mL. The present study's data clearly show that there is a considerable and clinically unacceptable lack of agreement between the S-FPIA and the EMIT techniques in HTx recipients for the whole range of concentrations evaluated (25-500 ng/mL), and this is caused by the variation in the overestimation of the CsA parent compound. Even though a similar CsA reference range was reported during maintenance therapy for both methods (150-250 ng/mL), which might encourage their interchangeability in the clinical setting, this approach should be avoided. Laboratory reports should always state both the concentration of CsA and the analytical method.


Asunto(s)
Ciclosporina/sangre , Monitoreo de Drogas/métodos , Trasplante de Corazón , Inmunosupresores/sangre , Adulto , Anciano , Anticuerpos Monoclonales , Especificidad de Anticuerpos , Técnica de Inmunoensayo de Enzimas Multiplicadas , Inmunoensayo de Polarización Fluorescente , Humanos , Persona de Mediana Edad , Reproducibilidad de los Resultados
18.
Int J Dev Biol ; 44(6): 699-706, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11061434

RESUMEN

In amniotes, myogenic commitment appears to be dependent upon signaling from neural tube and dorsal ectoderm, that can be replaced by members of the Wnt family and by Sonic hedgehog. Once committed, myoblasts undergo different fates, in that they can differentiate immediately to form the myotome, or later to give rise to primary and secondary muscle fibers. With fiber maturation, satellite cells are first detected; these cells contribute to fiber growth and regeneration during post-natal life. We will describe recent data, mainly from our laboratory, that suggest a different origin for some of the cells that are incorporated into the muscle fibers during late development. We propose the possibility that these myogenic cells are derived from the vasculature, are multi-potent and become committed to myogenesis by local signaling, when ingressing a differentiating muscle tissue. The implications for fetal and perinatal development of the whole mesoderm will also be discussed.


Asunto(s)
Linaje de la Célula , Mesodermo/metabolismo , Músculos/citología , Músculos/fisiología , Transactivadores , Proteínas de Pez Cebra , Animales , Diferenciación Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Proteínas Hedgehog , Ratones , Modelos Biológicos , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Wnt
20.
FASEB J ; 14(9): 1147-58, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10834937

RESUMEN

Transforming growth factor beta (TGF) is a well-known inhibitor of myogenic differentiation as well as an autocrine product of rhabdomyosarcoma cells. We studied the role of the TGF-beta autocrine loop in regulating growth and myogenic differentiation in the human rhabdomyosarcoma cell line, RD. We previously reported that the phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA) induces growth arrest and myogenic differentiation in these cells, which constitutively express muscle regulatory factors. We show that TPA inhibits the activation of secreted latent TGF-beta, thus decreasing the concentration of active TGF-beta to which the cells are exposed. This event is mediated by the TPA-induced alteration of the uPA/PAI serine-protease system. Complete removal of TGF-beta, mediated by the ectopic expression of a soluble type II TGF-beta receptor dominant negative cDNA, induces growth arrest, but does not trigger differentiation. In contrast, a reduction in the TGF-beta concentration, to a range of 0.14-0.20 x 10(-2) ng/ml (which is similar to that measured in TPA-treated cells), mimics TPA-induced differentiation. Taken together, these data demonstrate that cell growth and suppression of differentiation in rhabdomyosarcoma cells require overproduction of active TGF-beta; furthermore, they show that a 'critical' concentration of TGF-beta is necessary for myogenic differentiation to occur, whereas myogenesis is abolished below and above this concentration. By impairing the TGF-beta autocrine loop, TPA stabilizes the factor concentration within the range compatible for differentiation to occur. In contrast, in human primary muscle cells a much higher concentration of exogenous TGF-beta is required for the differentiation inhibitory effect and TPA inhibits differentiation in these cells probably through a TGF-beta independent mechanism. These data thus clarify the mechanism underlying the multiple roles of TGF-beta in the regulation of both the transformed and differentiated phenotype.


Asunto(s)
Comunicación Autocrina/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Músculo Esquelético/citología , Rabdomiosarcoma/patología , Factor de Crecimiento Transformador beta/farmacología , Animales , Aprotinina/farmacología , División Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutación/genética , Cadenas Pesadas de Miosina/metabolismo , Pepstatinas/farmacología , Plasminógeno/metabolismo , Inactivadores Plasminogénicos/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Rabdomiosarcoma/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Transfección , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA