Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
NMR Biomed ; 35(3): e4659, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34841594

RESUMEN

31 Phosphorus magnetic resonance spectroscopy (31 P-MRS) has been shown to detect altered energetic status (e.g. the ratio of inorganic phosphate to phosphocreatine: Pi/PCr), intracellular acid-base status, and free intracellular magnesium ([Mg2+ ]) in dystrophic muscle compared with unaffected muscle; however, the causes of these differences are not well understood. The purposes of this study were to examine 31 P-MRS indices of energetic status and sarcolemma integrity in young mdx mice compared with wild-type and to evaluate the effects of downhill running to induce muscle damage on 31 P-MRS indices in dystrophic muscle. In vivo 31 P-MRS spectra were acquired from the posterior hindlimb muscles in young (4-10 weeks of age) mdx (C57BL/10ScSn-DMDmdx) and wild-type (C57BL/10ScSnJ) mice using an 11.1-T MR system. The flux of phosphate from PCr to ATP was estimated by 31 P-MRS saturation transfer experiments. Relative concentrations of high-energy phosphates were measured, and intracellular pH and [Mg2+ ] were calculated. 1 H2 O-T2 was measured using single-voxel 1 H-MRS from the gastrocnemius and soleus using a 4.7-T MR system. Downhill treadmill running was performed in a subset of mice. Young mdx mice were characterized by elevated 1 H2 O-T2 (p < 0.01), Pi/PCr (p = 0.02), PCr to ATP flux (p = 0.04) and histological inflammatory markers (p < 0.05) and reduced (p < 0.01) [Mg2+ ] compared with wild-type. Furthermore, 24 h after downhill running, an increase (p = 0.02) in Pi/PCr was observed in mdx and wild-type mice compared with baseline, and a decrease (p < 0.001) in [Mg2+ ] and a lower (p = 0.048) intracellular [H+ ] in damaged muscle regions of mdx mice were observed, consistent with impaired sarcolemma integrity. Overall, our findings demonstrate that 31 P-MRS markers of energetic status and sarcolemma integrity are altered in young mdx compared with wild-type mice, and these indices are exacerbated following downhill running.


Asunto(s)
Metabolismo Energético , Distrofia Muscular Animal/metabolismo , Sarcolema/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Espectroscopía de Resonancia Magnética/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Fosfocreatina/metabolismo , Fósforo , Condicionamiento Físico Animal
2.
Photochem Photobiol ; 96(1): 200-207, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31733143

RESUMEN

This study analyzed photobiomodulation therapy (PBMT) effects on regenerative, antioxidative, anti-inflammatory and angiogenic markers in the dystrophic skeletal muscle of mdx mice, the experimental model of Duchenne muscular dystrophy (DMD), during the acute phase of dystrophy disease. The following groups were set up: Ctrl (control group of normal wild-type mice; C57BL/10); mdx (untreated mdx mice); mdxPred (mdx mice treated with prednisolone) and mdxLA (mdx mice treated with PBMT). The PBMT was carried out using an Aluminum Gallium Arsenide (AIGaAs; IBRAMED® laserpulse) diode, 830 nm wavelength, applied on the dystrophic quadriceps muscle. The mdxLA group showed a degenerative and regenerative area reduction simultaneously with a MyoD level increase, ROS production and inflammatory marker reduction and up-regulation in the VEGF factor. In addition, PBMT presented similar effects to prednisolone treatment in most of the parameters analyzed. In conclusion, our results indicate that PBMT in the parameters selected attenuated the dystrophic phenotype of mdx mice, improving skeletal muscle regeneration; reducing the oxidative stress and inflammatory process; and up-regulating the angiogenic marker.


Asunto(s)
Terapia por Luz de Baja Intensidad , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Fenotipo , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Sci Rep ; 9(1): 12982, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31506484

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked genetic disease characterized by progressive muscle wasting and weakness and premature death. Glucocorticoids (e.g. prednisolone) remain the only drugs with a favorable impact on DMD patients, but not without side effects. We have demonstrated that glycine preserves muscle in various wasting models. Since glycine effectively suppresses the activity of pro-inflammatory macrophages, we investigated the potential of glycine treatment to ameliorate the dystrophic pathology. Dystrophic mdx and dystrophin-utrophin null (dko) mice were treated with glycine or L-alanine (amino acid control) for up to 15 weeks and voluntary running distance (a quality of life marker and strong correlate of lifespan in dko mice) and muscle morphology were assessed. Glycine increased voluntary running distance in mdx mice by 90% (P < 0.05) after 2 weeks and by 60% (P < 0.01) in dko mice co-treated with prednisolone over an 8 week treatment period. Glycine treatment attenuated fibrotic deposition in the diaphragm by 28% (P < 0.05) after 10 weeks in mdx mice and by 22% (P < 0.02) after 14 weeks in dko mice. Glycine treatment augmented the prednisolone-induced reduction in fibrosis in diaphragm muscles of dko mice (23%, P < 0.05) after 8 weeks. Our findings provide strong evidence that glycine supplementation may be a safe, simple and effective adjuvant for improving the efficacy of prednisolone treatment and improving the quality of life for DMD patients.


Asunto(s)
Modelos Animales de Enfermedad , Glicinérgicos/administración & dosificación , Glicina/administración & dosificación , Distrofia Muscular Animal/tratamiento farmacológico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Prednisolona/farmacología , Animales , Antiinflamatorios/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Noqueados , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología
4.
Am J Physiol Heart Circ Physiol ; 312(1): H128-H140, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-27836895

RESUMEN

Duchenne Muscular Dystrophy (DMD) is associated with progressive cardiac pathology; however, the SIRT1/PGC1-α activator quercetin may cardioprotect dystrophic hearts. We tested the extent to which long-term 0.2% dietary quercetin enrichment attenuates dystrophic cardiopathology in Mdx/Utrn+/- mice. At 2 mo, Mdx/Utrn+/- mice were fed quercetin-enriched (Mdx/Utrn+/--Q) or control diet (Mdx/Utrn+/-) for 8 mo. Control C57BL/10 (C57) animals were fed a control diet for 10 mo. Cardiac function was quantified by MRI at 2 and 10 mo. Spontaneous physical activity was quantified during the last week of treatment. At 10 mo hearts were excised for histological and biochemical analysis. Quercetin feeding improved various physiological indexes of cardiac function in diseased animals. Mdx/Utrn+/--Q also engaged in more high-intensity physical activity than controls. Histological analyses of heart tissues revealed higher expression and colocalization of utrophin and α-sarcoglycan. Lower abundance of fibronectin, cardiac damage (Hematoxylin Eosin-Y), and MMP9 were observed in quercetin-fed vs. control Mdx/Utrn+/- mice. Quercetin evoked higher protein abundance of PGC-1α, cytochrome c, ETC complexes I-V, citrate synthase, SOD2, and GPX compared with control-fed Mdx/Utrn+/- Quercetin decreased abundance of inflammatory markers including NFκB, TGF-ß1, and F4/80 compared with Mdx/Utrn+/-; however, P-NFκB, P-IKBα, IKBα, CD64, and COX2 were similar between groups. Dietary quercetin enrichment improves cardiac function in aged Mdx/Utrn+/- mice and increases mitochondrial protein content and dystrophin glycoprotein complex formation. Histological analyses indicate a marked attenuation in pathological cardiac remodeling and indicate that long-term quercetin consumption benefits the dystrophic heart. NEW & NOTEWORTHY: The current investigation provides first-time evidence that quercetin provides physiological cardioprotection against dystrophic pathology and is associated with improved spontaneous physical activity. Secondary findings suggest that quercetin-dependent outcomes are in part due to PGC-1α pathway activation.


Asunto(s)
Antioxidantes/farmacología , Corazón/efectos de los fármacos , Distrofia Muscular Animal/fisiopatología , Quercetina/farmacología , Animales , Antígenos de Diferenciación/efectos de los fármacos , Antígenos de Diferenciación/metabolismo , Western Blotting , Citrato (si)-Sintasa/efectos de los fármacos , Citrato (si)-Sintasa/metabolismo , Ciclooxigenasa 2/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Citocromos c/efectos de los fármacos , Citocromos c/metabolismo , Modelos Animales de Enfermedad , Proteínas del Complejo de Cadena de Transporte de Electrón/efectos de los fármacos , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Fibronectinas/metabolismo , Alimentos Fortificados , Corazón/diagnóstico por imagen , Corazón/fisiopatología , Imagen por Resonancia Magnética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos mdx , Mitocondrias Musculares/efectos de los fármacos , Mitocondrias Musculares/metabolismo , Actividad Motora , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne , Miocardio/metabolismo , Miocardio/patología , Inhibidor NF-kappaB alfa/efectos de los fármacos , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Fosforilación , Receptores de IgG/efectos de los fármacos , Receptores de IgG/metabolismo , Sarcoglicanos/metabolismo , Superóxido Dismutasa/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Factor de Crecimiento Transformador beta1/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Utrofina/genética , Utrofina/metabolismo
5.
Amino Acids ; 48(11): 2635-2645, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27444300

RESUMEN

This study investigated the effect of taurine and ß-alanine supplementation on muscle function and muscle taurine transporter (TauT) protein expression in mdx mice. Wild-type (WT) and mdx mice (5 months) were supplemented with taurine or ß-alanine for 4 weeks, after which in vitro contractile properties, fatigue resistance and force recovery, and the expression of the TauT protein and proteins involved in excitation-contraction (E-C) coupling were examined in fast-twitch muscle. There was no difference in basal TauT protein expression or basal taurine content between mdx than WT muscle. Supplementation with taurine and ß-alanine increased and reduced taurine content, respectively, in muscle from WT and mdx mice but had no effect of TauT protein. Taurine supplementation reduced body and muscle mass, and enhanced fatigue resistance and force recovery in mdx muscle. ß-Alanine supplementation enhanced fatigue resistance in WT and mdx muscle. There was no difference in the basal expression of key E-C coupling proteins [ryanodine receptor 1 (RyR1), dihydropyridine receptor (DHPR), sarco(endo)plasmic reticulum Ca2+-ATPase 1 (SERCA1) or calsequestrin 1 (CSQ1)] between WT and mdx mice, and the expression of these proteins was not altered by taurine or ß-alanine supplementation. These findings suggest that TauT protein expression is relatively insensitive to changes in muscle taurine content in WT and mdx mice, and that taurine and ß-alanine supplementation may be viable therapeutic strategies to improve fatigue resistance of dystrophic skeletal muscle.


Asunto(s)
Fatiga/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas de Membrana/biosíntesis , Proteínas de Transporte de Membrana/biosíntesis , Músculo Esquelético/metabolismo , Taurina/farmacología , beta-Alanina/farmacología , Animales , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/patología , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología
6.
Free Radic Biol Med ; 83: 129-38, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25668720

RESUMEN

Nutritional muscular dystrophy (NMD) of chicks is induced by dietary selenium (Se)/vitamin E (Vit. E) deficiencies and may be associated with oxidative cell damage. To reveal the underlying mechanisms related to the presumed oxidative cell damage, we fed four groups of 1-day-old broiler chicks (n = 40/group) with a basal diet (BD; 10 µg Se/kg; no Vit. E added, -Se -Vit. E) or the BD plus all-rac-α-tocopheryl acetate at 50mg/kg (-Se +Vit. E), Se (as sodium selenite) at 0.3mg/kg (+Se -Vit. E), or both of these nutrients (+Se +Vit. E) for 6 weeks. High incidences of NMD (93%) and mortality (36%) of the chicks were induced by the BD, starting at week 3. Dietary Se deficiency alone also induced muscle fiber rupture and coagulation necrosis in the pectoral muscle of chicks at week 3 and thereafter, with increased (P < 0.05) malondialdehyde, decreased (P < 0.05) total antioxidant capacity, and diminished (P < 0.05) glutathione peroxidase activities in the muscle. To link these oxidative damages of the muscle cells to the Se-deficiency-induced NMD, we first determined gene expression of the potential 26 selenoproteins in the muscle of the chicks at week 2 before the onset of symptoms. Compared with the +Se chicks, the -Se chicks had lower (P < 0.05) muscle mRNA levels of Gpx1, Gpx3, Gpx4, Sepp1, Selo, Selk, Selu, Selh, Selm, Sepw1, and Sep15. The -Se chicks also had decreased (P < 0.05) production of 6 selenoproteins (long-form selenoprotein P (SelP-L), GPx1, GPx4, Sep15, SelW, and SelN), but increased levels (P < 0.05) of the short-form selenoprotein P in muscle at weeks 2 and 4. Dietary Se deficiency elevated (P < 0.05) muscle p53, cleaved caspase 3, cleaved caspase 9, cyclooxygenase 2 (COX2), focal adhesion kinase (FAK), phosphatidylinositol 3-kinase (PI3K), phospho-Akt, nuclear factor-κB (NF-κB), p38 mitogen-activated protein kinase (p38 MAPK), phospho-p38 MAPK, phospho-JNK, and phospho-ERK and decreased (P < 0.05) muscle procaspase 3, procaspase 9, and NF-κB inhibitor α. In conclusion, the downregulation of SelP-L, GPx1, GPx4, Sep15, SelW, and SelN by dietary Se deficiency might account for induced oxidative stress and the subsequent peroxidative damage of chick muscle cells via the activation of the p53/caspase 9/caspase 3, COX2/FAK/PI3K/Akt/NF-κB, and p38 MAPK/JNK/ERK signaling pathways. Metabolism of peroxides and redox regulation are likely to be the mechanisms whereby these selenoproteins prevented the onset of NMD in chicks.


Asunto(s)
Apoptosis , Dieta/efectos adversos , Distrofia Muscular Animal/prevención & control , Peróxidos/metabolismo , Selenoproteínas/metabolismo , Animales , Antioxidantes , Western Blotting , Proliferación Celular , Células Cultivadas , Pollos , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Técnicas para Inmunoenzimas , Masculino , Distrofia Muscular Animal/etiología , Distrofia Muscular Animal/metabolismo , Oxidación-Reducción , Estrés Oxidativo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Selenio/metabolismo , Selenoproteínas/genética , Glutatión Peroxidasa GPX1
7.
Exp Physiol ; 100(1): 12-22, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25557727

RESUMEN

NEW FINDINGS: What is the central question of this study? Does dietary quercetin enrichment improve biochemical and histological outcomes in hearts from mdx mice? What is the main finding and what is its importance? Biochemical and histological findings suggest that chronic quercetin feeding of mdx mice may improve mitochondrial function and attenuate tissue pathology. Patients with Duchenne muscular dystrophy suffer from cardiac pathology, which causes up to 40% of all deaths because of fibrosis and cardiac complications. Quercetin is a flavonol with anti-inflammatory and antioxidant effects and is also an activator of peroxisome proliferator-activated receptor γ coactivator 1α capable of antioxidant upregulation, mitochondrial biogenesis and prevention of cardiac complications. We sought to determine the extent to which dietary quercetin enrichment prevents (experiment 1) and rescues cardiac pathology (experiment 2) in mdx mice. In experiment 1, 3-week-old mdx mice were fed control chow (C3w6m, n = 10) or chow containing 0.2% quercetin for 6 months (Q3w6m, n = 10). In experiment 2, 3-month-old mdx mice were fed control chow (C3m6m, n = 10) or 0.2% chow containing 0.2% quercetin for 6 months (Q3m6m, n = 10). Hearts were excised for histological and biochemical analyses. In experiment 1, Western blot targets for mitochondrial biogenesis (cytochrome c, P = 0.007) and antioxidant expression (superoxide dismutase 2, P = 0.014) increased in Q3w6m mice compared with C3w6m. Histology revealed increased utrophin (P = 0.025) and decreased matrix metalloproteinase 9 abundance (P = 0.040) in Q3w6m mice compared with C3w6m. In experiment 2, relative (P = 0.023) and absolute heart weights (P = 0.020) decreased in Q3m6m mice compared with C3m6m. Indications of damage (Haematoxylin- and Eosin-stained sections, P = 0.007) and Western blot analysis of transforming growth factor ß1 (P = 0.009) were decreased in Q3m6m mice. Six months of quercetin feeding increased a mitochondrial biomarker, antioxidant protein and utrophin and decreased matrix metalloproteinase 9 in young mice. Given that these adaptations are associated with attenuated cardiac pathology and damage, the present findings may indicate that dietary quercetin enrichment attenuates dystrophic cardiac pathology, but physiological confirmation is needed.


Asunto(s)
Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/prevención & control , Suplementos Dietéticos , Mitocondrias Cardíacas/efectos de los fármacos , Distrofia Muscular Animal/tratamiento farmacológico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Miocardio/patología , Quercetina/farmacología , Animales , Cardiomiopatías/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Citocromos c/metabolismo , Citoprotección , Modelos Animales de Enfermedad , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos mdx , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Recambio Mitocondrial/efectos de los fármacos , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Miocardio/metabolismo , Superóxido Dismutasa/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta1/metabolismo , Utrofina/metabolismo
8.
PLoS One ; 8(7): e66617, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23843959

RESUMEN

BACKGROUND: In Duchenne muscular dystrophy (DMD), loss of the membrane stabilizing protein dystrophin results in myofiber damage. Microinjury to dystrophic myofibers also causes secondary imbalances in sarcolemmic ion permeability and resting membrane potential, which modifies excitation-contraction coupling and increases proinflammatory/apoptotic signaling cascades. Although glucocorticoids remain the standard of care for the treatment of DMD, there is a need to investigate the efficacy of other pharmacological agents targeting the involvement of imbalances in ion flux on dystrophic pathology. METHODOLOGY/PRINCIPAL FINDINGS: We designed a preclinical trial to investigate the effects of lansoprazole (LANZO) administration, a proton pump inhibitor, on the dystrophic muscle phenotype in dystrophin deficient (mdx) mice. Eight to ten week-old female mice were assigned to one of four treatment groups (n = 12 per group): (1) vehicle control; (2) 5 mg/kg/day LANZO; (3) 5 mg/kg/day prednisolone; and (4) combined treatment of 5 mg/kg/day prednisolone (PRED) and 5 mg/kg/day LANZO. Treatment was administered orally 5 d/wk for 3 months. At the end of the study, behavioral (Digiscan) and functional outcomes (grip strength and Rotarod) were assessed prior to sacrifice. After sacrifice, body, tissue and organ masses, muscle histology, in vitro muscle force, and creatine kinase levels were measured. Mice in the combined treatment groups displayed significant reductions in the number of degenerating muscle fibers and number of inflammatory foci per muscle field relative to vehicle control. Additionally, mice in the combined treatment group displayed less of a decline in normalized forelimb and hindlimb grip strength and declines in in vitro EDL force after repeated eccentric contractions. CONCLUSIONS/SIGNIFICANCE: Together our findings suggest that combined treatment of LANZO and prednisolone attenuates some components of dystrophic pathology in mdx mice. Our findings warrant future investigation of the clinical efficacy of LANZO and prednisolone combined treatment regimens in dystrophic pathology.


Asunto(s)
Distrofina/genética , Lansoprazol/farmacología , Fuerza Muscular/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular Animal/tratamiento farmacológico , Inhibidores de la Bomba de Protones/farmacología , Animales , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Distrofina/deficiencia , Femenino , Expresión Génica , Glucocorticoides/farmacología , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Prednisolona/farmacología
9.
Neuromuscul Disord ; 22(1): 43-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21737275

RESUMEN

Duchenne Muscular Dystrophy is an X-linked disorder that affects boys and leads to muscle wasting and death due to cardiac involvement and respiratory complications. The cause is the absence of dystrophin, a large structural protein indispensable for muscle cell function and viability. The mdx mouse has become the standard animal model for pre-clinical evaluation of potential therapeutic treatments. Recent years have seen a rapid increase in the number of experimental compounds being evaluated in the mdx mouse. There is, however, much variability in the design of these pre-clinical experimental studies. This has made it difficult to interpret and compare published data from different laboratories and to evaluate the potential of a treatment for application to patients. The authors therefore propose the introduction of a standard study design for the mdx mouse model. Several aspects, including animal care, sampling times and choice of tissues, as well as recommended endpoints and methodologies are addressed and, for each aspect, a standard procedure is proposed. Testing of all new molecules/drugs using a widely accepted and agreed upon standard experimental protocol would greatly improve the power of pre-clinical experimentations and help identifying promising therapies for the translation into clinical trials for boys with Duchenne Muscular Dystrophy.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Endogámicos mdx , Distrofia Muscular de Duchenne/tratamiento farmacológico , Animales , Evaluación Preclínica de Medicamentos/normas , Distrofina/normas , Humanos , Masculino , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/diagnóstico , Distrofia Muscular Animal/tratamiento farmacológico , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/diagnóstico , Distrofia Muscular de Duchenne/metabolismo
10.
J Cell Biochem ; 111(6): 1652-60, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21053281

RESUMEN

Eugenia punicifolia known as "pedra-ume caá" is a shrub largely distributed in the Amazon region popularly used in decoctions or infusions as a natural therapeutic agent, which can interfere on cholinergic nicotinic neurotransmission. This work aimed to investigate a putative anti-inflammatory effect of dichloromethane fraction of E. punicifolia extract (Ep-CM) in the muscular lesion of mdx dystrophic mice, considering that activation of cholinergic mechanisms mitigates inflammation. A polymer containing the Ep-CM was implanted in mdx gastrocnemius muscle before onset of myonecrosis for local slow and gradual release of bioactive compounds and mice sacrificed 7 days or 9 weeks after surgery. Comparing to control muscle, treatment did not alter choline acetyltransferase and acetylcholinesterase enzymatic activities, but decreased metaloproteases-9 and -2 activities and levels of tumor necrosis factor α and NFκB transcription factor. In addition, treatment also reduced levels of bioactive IL-1ß form and cleaved caspase-3, related to early events of cellular death and inflammatory activation and further increased myogenin expression without affecting collagen production which is associated with fibrosis. In vivo treatment of mdx dystrophic mice with Ep-CM caused significant reduction of muscular inflammation and improved skeletal muscle regeneration without inducing fibrosis.


Asunto(s)
Antiinflamatorios/uso terapéutico , Distrofia Muscular Animal/tratamiento farmacológico , Extractos Vegetales/uso terapéutico , Syzygium/química , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Colina O-Acetiltransferasa/metabolismo , Interleucina-1beta/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/efectos de los fármacos , Distrofia Muscular Animal/metabolismo , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
11.
FEBS Lett ; 439(3): 357-62, 1998 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-9845353

RESUMEN

Dystrophic skeletal muscle cells from Duchenne muscular dystrophy (DMD) patients and mdx mice exhibit elevated cytosolic Ca2+ concentrations ([Ca2+]c). Pretreatment of mdr myotubes for 6-12 days with creatine (20 mM) decreased the elevation in [Ca2+]c induced by either high extracellular Ca2+ concentrations or hypo-osmotic stress to control levels. 45Ca2+ influx measurements suggest that creatine lowered [Ca2+]c by stimulating sarcoplasmic reticulum Ca2+-ATPase. Creatine pretreatment increased levels of phosphocreatine but not ATP. Furthermore, myotube formation and survival were significantly enhanced by creatine pretreatment. Therefore, creatine supplementation may be useful for treatment of DMD.


Asunto(s)
Calcio/metabolismo , Creatina/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Creatina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Distrofia Muscular Animal/genética , Presión Osmótica/efectos de los fármacos , Fosfocreatina/metabolismo , Factores de Tiempo
12.
Biochem Pharmacol ; 56(6): 751-7, 1998 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9751080

RESUMEN

Duchenne muscular dystrophy (DMD) is caused by dystrophin deficiency, which results in muscle necrosis and the upregulation of heat shock/stress proteins (HSP). We hypothesized that reactive oxygen species, and in particular hydroxyl radicals (.OH), participate in muscle necrosis and HSP expression. It was assumed that iron deprivation decreases .OH generation, restraining the disease process and reducing the oxidant-induced expression of HSP. The role of iron-catalyzed free radical reactions in the pathology of dystrophin-deficient muscle was evaluated in the murine model for Duchenne muscular dystrophy (mdx), by examining the effects of dietary deficiency and supplementation of iron on serum creatine kinase (CK), muscle morphology, lipid peroxidation and HSP levels in mice maintained on diets deficient in or supplemented with iron for 6 weeks. Iron-deprived mdx mice showed a significant decrease in the number of macrophage-invaded necrotic fibers and the expression of the 70-kDa heat shock protein (Hsp70). This suggests that the iron-dependent generation of .OH relates to muscle necrosis in the mdx mouse and modulates the expression of Hsp70 in vivo. In contrast, iron deprivation had no influence on other HSP or on lipid peroxidation in mdx mice, while maintenance on either diet caused a significant decrease in serum creatine kinase activity. The potential therapeutic effects of iron deprivation in mdx should be considered.


Asunto(s)
Proteínas de Choque Térmico/biosíntesis , Deficiencias de Hierro , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Animales , Creatina Quinasa/sangre , Suplementos Dietéticos , Modelos Animales de Enfermedad , Ligamiento Genético , Hierro/sangre , Hierro de la Dieta/administración & dosificación , Peroxidación de Lípido/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/patología , Distrofia Muscular Animal/genética , Necrosis , Cromosoma X
13.
Neuromuscul Disord ; 8(6): 371-9, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9713853

RESUMEN

31P NMR spectroscopy was used to study the energy metabolism of dystrophin-deficient skeletal muscle of mdx mice, an animal model of Duchenne muscular dystrophy, in which expression of a truncated form of utrophin has been obtained through transgenesis technology. Measurements of ATP, phosphocreatine (PCr), inorganic phosphates (Pi) and intracellular pH (pHi) were made at rest, during a fatigue protocol and during the subsequent recovery. Mechanical fatigue of transgenic muscles was similar to normal muscle, while mdx muscle showed larger force loss. At rest, muscles of all groups had similar values for [ATP], [PCr], [Pi] and pHi. During fatigue, [PCr] decreases mirrored [Pi] increases and were similar in all groups. The major difference between mdx muscles and the group of normal and trc-utrophin muscles concerned the values and evolution of pHi. The mdx muscles showed a more severe intracellular acidosis during exercise and a slower and incomplete post-exercise recovery of normal pHi. In contrast, in trc-utrophin muscles, the kinetics and amplitude of pHi changes were remarkably close to normal behaviour. We conclude that the impaired proton washout which is present in mdx muscles, is corrected to a great extent by the expression of trc-utrophin.


Asunto(s)
Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Hidrógeno/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Actividad Motora/fisiología , Distrofia Muscular Animal/fisiopatología , Animales , Concentración de Iones de Hidrógeno , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx/metabolismo , Ratones Endogámicos mdx/fisiología , Ratones Transgénicos , Distrofia Muscular Animal/diagnóstico , Distrofia Muscular Animal/metabolismo , Fósforo , Utrofina
14.
Biochim Biophys Acta ; 1362(2-3): 201-7, 1997 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-9540851

RESUMEN

The expression of isoform-specific dihydropyrine receptor-calcium channel (DHPR) alpha 1-subunit genes was investigated in mdx and control mouse diaphragm (DIA) and tibialis anterior (TA). RNase protection assays were carried out with a rat DHPR cDNA probe specific for skeletal muscle and a mouse DHPR cDNA probe specific for cardiac muscle. The level of expression of the gene encoding the cardiac DHPR was very weak in TA muscle from both control and mdx mice. Compared to TA, DIA expressed mRNA for the cardiac isoform at significantly higher levels, but mdx and control mouse DIA levels were similar to one another. In contrast, mRNA expression levels for the DHPR skeletal muscle isoform were lower in control DIA than TA. However, there was a dramatic increase in the expression for the DHPR skeletal muscle isoform in mdx DIA compared with control DIA, reaching the TA expression level, whereas dystrophy did not affect TA expression. [3H]-PN200-110 binding was used to further assess DIA DHPR expression at the protein level. The density of binding sites for the probe was not significantly affected in DIA muscles of mdx vs. control mice, but it was reduced in older mdx and control mice. The increase in DHPR mRNA levels without a consequent increase in DHPR protein expression could be secondary to possible enhanced protein degradation which occurs in mdx DIA. The altered DHPR expression levels found here do not appear to be responsible for the severe deficits in contractile function of the mdx DIA.


Asunto(s)
Canales de Calcio/genética , Distrofia Muscular Animal/genética , Animales , Canales de Calcio/metabolismo , Canales de Calcio Tipo L , ADN Complementario/genética , Expresión Génica , Isradipino/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo , Miocardio/metabolismo , ARN Mensajero/genética
15.
Endocrinology ; 136(11): 4880-6, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7588220

RESUMEN

In muscular dystrophy there is an imbalance between muscle protein synthesis and protein degradation, resulting in net muscle catabolism and progressive muscle weakness and wasting. Both insulin and insulin-like growth factor I (IGF-I) are known to have an anabolic effect on skeletal muscle, which is believed to be enhanced in the presence of elevated concentrations of amino acids. We examined the effects of 4-week administration of recombinant human IGF-I (rhIGF-I), both alone and supplemented with a high protein diet (HPD), on muscle metabolism, morphology, and function in the 129 ReJ dystrophic mouse. rhIGF-I significantly reduced muscle protein degradation (P < 0.001), increased muscle protein content (P < 0.05), decreased fiber area variability (P < 0.01), and increased hind limb utilization (P < 0.01). Supplementation of rhIGF-I therapy with a HPD resulted in a significant increase in muscle protein synthesis (P < 0.05) in addition to a further increase in the above parameters. We conclude that rhIGF-I causes an improvement in muscle metabolism, morphology, and function in dystrophic mice, and this effect is further enhanced by the presence of a HPD.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/farmacología , Proteínas Musculares/metabolismo , Distrofia Muscular Animal/metabolismo , Animales , Proteínas en la Dieta/administración & dosificación , Humanos , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Cinética , Masculino , Metilhistidinas/orina , Ratones , Proteínas Musculares/biosíntesis , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/tratamiento farmacológico , Distrofia Muscular Animal/fisiopatología , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Tirosina/metabolismo
16.
J Neurol Sci ; 133(1-2): 16-23, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8583220

RESUMEN

Mechanical properties and metabolic adaptation to exercise in skeletal muscle of dystrophic hamsters were studied with an in vivo 31P-NMR multistep fatigue test. Three successive 20-min steps with increasing rhythms of tetanic stimulation were followed by a 20-min recovery period. Fatigue in dystrophic hamsters (DH) developed more rapidly and was greater than in normal hamsters (NH); total mechanical performance per min increased step by step in NH while it decreased in DH, showing a progressive mechanical impairment of the dystrophic muscles. ADP and PCr recovery rates were significantly reduced in DH muscles. Acidosis appeared in both DH and NH and persisted in DH throughout the test, suggesting reduced mitochondrial oxidative capacity of the dystrophic muscle. The pH recovery rate was reduced in DH muscles suggesting a reduction in export protons capacity. These results provide evidence of impaired mitochondrial function and intracellular ionic regulation in the dystrophic muscle, associated with the lack of dystrophin and dystrophin-associated glycoproteins in the DH.


Asunto(s)
Metabolismo Energético/fisiología , Fatiga Muscular/fisiología , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/fisiopatología , Condicionamiento Físico Animal/fisiología , Análisis de Varianza , Animales , Fenómenos Biomecánicos , Cricetinae , Concentración de Iones de Hidrógeno , Espectroscopía de Resonancia Magnética , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo , Oxidación-Reducción , Fosfocreatina/metabolismo , Fósforo
17.
Proc Biol Sci ; 251(1332): 201-6, 1993 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-8097327

RESUMEN

Intracellular pH, ratios of phosphocreatine (PCr) to ATP and PCr to inorganic phosphate (Pi) as well as isometric tension were measured during 1 Hz sciatic nerve stimulation and during recovery in the calf muscles of mdx (a model of Duchenne muscular dystrophy) and control mice. Tension did not decline significantly in either strain. The ratio of PCr/(PCr + Pi) was significantly reduced in mdx as against control muscle during exercise and recovery, but the ratio of PCr/ATP and the half-time for PCr recovery were similar in both strains. A reduction in the maximal activities of succinate dehydrogenase and succinate-cytochrome c reductase suggests that mitochondrial metabolism may be impaired. The similarity in PCr recovery times suggests that the muscle has adapted, making any impairment of oxidative metabolism negligible in the intact system. The rate of pH recovery is prolonged in mdx muscle and provides strong evidence for a decline in the capacity of dystrophic muscle to extrude proton equivalents. These data are compared with a previous study which used 10 Hz stimulation and also observed a slow pH recovery. The slow pH recovery could be explained by an elevation in intracellular sodium.


Asunto(s)
Metabolismo Energético , Músculos/metabolismo , Distrofia Muscular Animal/metabolismo , Nervio Ciático/fisiopatología , Adenosina Trifosfato/metabolismo , Animales , Estimulación Eléctrica , Contracción Isométrica , Espectroscopía de Resonancia Magnética/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Músculos/inervación , Músculos/fisiopatología , Distrofia Muscular Animal/fisiopatología , Fosfatos/metabolismo , Fosfocreatina/metabolismo , Fósforo , Esfuerzo Físico
18.
J Neurol Sci ; 113(1): 108-13, 1992 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-1469448

RESUMEN

We have studied exercise metabolism in vivo in the mdx mouse model of Duchenne muscular dystrophy with 31P-nuclear magnetic resonance spectroscopy. Intracellular pH, ratios of phosphocreatine (PCr) to ATP and PCr to inorganic phosphate (P(i)) expressed as PCr/ATP and PCr/(PCr+P(i)) as well as tension generated at the Achilles tendon were measured during sciatic nerve stimulation. Tension was similar between the mdx and control strain C57Bl/10ScSn at 10 Hz stimulation but slightly higher than the control at 100 Hz. The PCr/ATP and PCr/(PCr+P(i)) ratios were significantly reduced in mdx vs. control muscle during exercise. Although resting muscle pH in mdx mice is more alkaline than normal muscle, the pH of mdx muscle during exercise is reduced relative to controls, as is the rate of pH recovery. Total lactate is not elevated in the cells and so it is argued that there is a reduction in the capacity to export proton equivalents in muscles of mdx mice which could be caused by an elevation in intracellular sodium. This provides more evidence of impaired ionic regulation in dystrophic muscle and could be used as an index for the evaluation in vivo of therapeutic interventions such as myoblast transfer or gene replacement therapy.


Asunto(s)
Espectroscopía de Resonancia Magnética , Músculos/metabolismo , Distrofia Muscular Animal/metabolismo , Esfuerzo Físico , Adenosina Trifosfato/metabolismo , Animales , Estimulación Eléctrica , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos C57BL , Contracción Muscular , Fósforo , Tiempo de Reacción
19.
Biochem Cell Biol ; 69(12): 835-41, 1991 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1667976

RESUMEN

Phospholipid incorporation of 32P by primary myotube cultures and the tissue activity of sarcolemmal Na+/K(+)-transporting ATPase were studied to determine whether the absence of dystrophin from dystrophic (mdx) muscle would affect membrane lipid synthesis and membrane function. The incorporation of 32P by phospholipid as a ratio with total protein was greater in cultured dystrophic cells compared with control cells. The mdx cells also incorporated more 32P than control cells into phosphatidylethanolamine, which is thought to increase prior to myoblast fusion, and less into phosphatidylserine, phosphatidylinositol, and lysophosphatidylcholine. There was no difference in total protein content or [3H]leucine or 32P incorporation into the aqueous fraction of dystrophic and control cells, although dystrophic cells incorporated less [35S]methionine into protein than controls. Isolated sarcolemma from mdx skeletal muscle tissue demonstrated a consistently greater specific activity of ouabain-sensitive Na+/K(+)-transporting ATPase than sarcolemmal preparations from control skeletal muscle. These observations suggest that cytoskeletal changes such as dystrophin deficiency may alter the differentiation of membrane composition and function.


Asunto(s)
Músculos/metabolismo , Distrofia Muscular Animal/metabolismo , Fosfolípidos/biosíntesis , Sarcolema/enzimología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , ATPasa de Ca(2+) y Mg(2+)/metabolismo , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/biosíntesis , Músculos/citología , Distrofia Muscular Animal/enzimología , Fósforo/metabolismo
20.
Biol Trace Elem Res ; 24(2): 163-71, 1990 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-1702668

RESUMEN

Selenium deficiency is responsible for Zenker type muscle degeneration in calves, lambs, and foals in the prenatal and postnatal stages of development. Investigations have shown that the selenium GSH Px, and vitamin E content of the maternal and fetal parts of the placenta in cattle are different. Similarly, low concentrations of selenium are present in milk from cows and sheep. In addition to an inadequate supply of selenium and vitamin E as a contributory cause of fetal nutritive muscular dystrophy (FNMD), it is assumed that a placental transport block and/or impaired selenium metabolism in the placenta are also responsible. Postnatal nutritive muscular dystrophy, however, is attributed to either acute selenium and vitamin E deficiency in basic feed or impaired plant absorption of selenium as a result of antagonistic elements, such as sulphur.


Asunto(s)
Enfermedades de los Bovinos/metabolismo , Intercambio Materno-Fetal , Distrofia Muscular Animal/metabolismo , Selenio/deficiencia , Enfermedades de las Ovejas/metabolismo , Animales , Animales Recién Nacidos , Bovinos , Femenino , Leche/metabolismo , Necesidades Nutricionales , Embarazo , Selenio/sangre , Ovinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA