Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 12: 766112, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34938290

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global health concern. The development of vaccines with high immunogenicity and safety is crucial for controlling the global COVID-19 pandemic and preventing further illness and fatalities. Here, we report the development of a SARS-CoV-2 vaccine candidate, Nanocovax, based on recombinant protein production of the extracellular (soluble) portion of the spike (S) protein of SARS-CoV-2. The results showed that Nanocovax induced high levels of S protein-specific IgG and neutralizing antibodies in three animal models: BALB/c mouse, Syrian hamster, and a non-human primate (Macaca leonina). In addition, a viral challenge study using the hamster model showed that Nanocovax protected the upper respiratory tract from SARS-CoV-2 infection. Nanocovax did not induce any adverse effects in mice (Mus musculus var. albino) and rats (Rattus norvegicus). These preclinical results indicate that Nanocovax is safe and effective.


Asunto(s)
Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/toxicidad , COVID-19/prevención & control , Inmunogenicidad Vacunal/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Cricetinae , Macaca , Ratones , Ratas , SARS-CoV-2 , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/toxicidad
2.
Annu Int Conf IEEE Eng Med Biol Soc ; 2019: 2848-2851, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31946486

RESUMEN

Cortical thickness measurement estimated from high-resolution anatomical MRI scans may serve as a marker of cortical atrophy in clinical research applications. Most of the working algorithms and pipelines are optimized for human in-vivo data analyses that offer robust and reproducible measures. As animal-models are widely utilized in many preclinical phases of clinical trials the need for an optimized automated MRI data analysis to yield reliable data is warranted. We present a processing pipeline optimized for cortical thickness estimation of canine brains in native and template spaces. Preliminary results of 5 healthy and 5 mucopolysaccharidosis (MPS) dogs demonstrate single-canine mean/median cortical thickness in range of 2.69-3.58mm in native space and 3.26-4.15mm in template space. Our MRI generated values exceed previous histological measurements (observed mean about 2mm) in limited literature reports. Randomly selected manual measures corroborated the ranges defined by estimated cortical thickness probability density functions. Geometric transformations between native and template spaces change absolute mean/median cortical thickness values, but do not change the data nature and properties since the Pearson correlation coefficients between different space estimates were 0.84 for mean values and 0.89 for median values. No significant difference in total cortical thickness between MPS and age-and gender-matched dogs was observed.


Asunto(s)
Enfermedades de los Perros , Imagen por Resonancia Magnética , Mucopolisacaridosis , Algoritmos , Animales , Atrofia , Encéfalo , Corteza Cerebral , Enfermedades de los Perros/diagnóstico por imagen , Perros , Imagen por Resonancia Magnética/veterinaria , Mucopolisacaridosis/diagnóstico por imagen , Mucopolisacaridosis/veterinaria
3.
Stem Cells ; 35(7): 1815-1834, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28480592

RESUMEN

Recently, we found that resident myogenic stem satellite cells upregulate a multi-functional secreted protein, semaphorin 3A (Sema3A), exclusively at the early-differentiation phase in response to muscle injury; however, its physiological significance is still unknown. Here we show that Sema3A impacts slow-twitch fiber generation through a signaling pathway, cell-membrane receptor (neuropilin2-plexinA3) → myogenin-myocyte enhancer factor 2D → slow myosin heavy chain. This novel axis was found by small interfering RNA-transfection experiments in myoblast cultures, which also revealed an additional element that Sema3A-neuropilin1/plexinA1, A2 may enhance slow-fiber formation by activating signals that inhibit fast-myosin expression. Importantly, satellite cell-specific Sema3A conditional-knockout adult mice (Pax7CreERT2 -Sema3Afl °x activated by tamoxifen-i.p. injection) provided direct in vivo evidence for the Sema3A-driven program, by showing that slow-fiber generation and muscle endurance were diminished after repair from cardiotoxin-injury of gastrocnemius muscle. Overall, the findings highlight an active role for satellite cell-secreted Sema3A ligand as a key "commitment factor" for the slow-fiber population during muscle regeneration. Results extend our understanding of the myogenic stem-cell strategy that regulates fiber-type differentiation and is responsible for skeletal muscle contractility, energy metabolism, fatigue resistance, and its susceptibility to aging and disease. Stem Cells 2017;35:1815-1834.


Asunto(s)
Fibras Musculares de Contracción Lenta/metabolismo , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Regeneración/genética , Células Satélite del Músculo Esquelético/metabolismo , Semaforina-3A/genética , Animales , Cardiotoxinas/administración & dosificación , Diferenciación Celular , Regulación de la Expresión Génica , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibras Musculares de Contracción Lenta/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/lesiones , Mioblastos/citología , Mioblastos/efectos de los fármacos , Miogenina/genética , Miogenina/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuropilina-2/genética , Neuropilina-2/metabolismo , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Regeneración/efectos de los fármacos , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/efectos de los fármacos , Semaforina-3A/antagonistas & inhibidores , Semaforina-3A/metabolismo , Transducción de Señal , Tamoxifeno/farmacología
4.
Biol Rev Camb Philos Soc ; 92(3): 1389-1405, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27296513

RESUMEN

Current research on skeletal muscle injury and regeneration highlights the crucial role of nerve-muscle interaction in the restoration of innervation during that process. Activities of muscle satellite or stem cells, recognized as the 'currency' of myogenic repair, have a pivotal role in these events, as shown by ongoing research. More recent investigation of myogenic signalling events reveals intriguing roles for semaphorin3A (Sema3A), secreted by activated satellite cells, in the muscle environment during development and regeneration. For example, Sema3A makes important contributions to regulating the formation of blood vessels, balancing bone formation and bone remodelling, and inflammation, and was recently implicated in the establishment of fibre-type distribution through effects on myosin heavy chain gene expression. This review highlights the active or potential contributions of satellite-cell-derived Sema3A to regulation of the processes of motor neurite ingrowth into a regenerating muscle bed. Successful restoration of functional innervation during muscle repair is essential; this review emphasizes the integrative role of satellite-cell biology in the progressive coordination of adaptive cellular and tissue responses during the injury-repair process in voluntary muscle.


Asunto(s)
Músculo Esquelético/fisiología , Unión Neuromuscular/fisiología , Regeneración/fisiología , Semaforina-3A/metabolismo , Diferenciación Celular , Humanos , Músculo Esquelético/citología , Neuritas/fisiología
5.
Anim Sci J ; 88(3): 489-499, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27417667

RESUMEN

Our previous studies demonstrated that an 8-week intake of 5% (w/w) apple polyphenol (APP) in the diet improves muscle endurance of young-adult rats. In order to identify a lower limit of the dietary contribution of APP to the effect, the experiments were designed for lower-dose supplementation (8-week feeding of 0.5% APP in AIN-93G diet) to 12-week-old male Sprague-Dawley rats. Results clearly showed that the 0.5% APP diet significantly up-regulates slower myosin-heavy-chain (MyHC) isoform ratios (IIx and IIa relative to total MyHC) and myoglobin expression in lower hind-limb muscles examined (P < 0.05). There was a trend to increased fatigue resistance detected from measurements of relative isometric plantar-flexion force torque generated by a stimulus train delivered to the tibial nerve (F(98, 1372) = 1.246, P = 0.0574). Importantly, there was no significant difference in the animal body-phenotypes or locomotor activity shown as total moving distance in light and dark periods. Therefore, the present study encourages the notion that even low APP-intake may increase the proportions of fatigue-resistant myofibers, and has promise as a strategy for modifying performance in human sports and improving function in age-related muscle atrophy.


Asunto(s)
Suplementos Dietéticos , Malus , Fibras Musculares de Contracción Rápida/metabolismo , Polifenoles/administración & dosificación , Polifenoles/farmacología , Animales , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Fatiga Muscular/efectos de los fármacos , Mioglobina/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Isoformas de Proteínas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos
6.
Physiol Rep ; 3(9)2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26381016

RESUMEN

Regenerative mechanisms that regulate intramuscular motor innervation are thought to reside in the spatiotemporal expression of axon-guidance molecules. Our previous studies proposed an unexplored role of resident myogenic stem cell (satellite cell)-derived myoblasts as a key presenter of a secreted neural chemorepellent semaphorin 3A (Sema3A); hepatocyte growth factor (HGF) and basic fibroblast growth factor (FGF2) triggered its expression exclusively at the early differentiation phase. In order to advance this concept, the present study described that transmembrane heparan/chondroitin sulfate proteoglycans syndecan-2, 4 may be the plausible receptor candidates for HGF and FGF2 to signal Sema3A expression. Results showed that mRNA expression of syndecan-2, 4 was abundant (two magnitudes higher than syndecan-1, 3) in early-differentiated myoblasts and their in vitro knockdown diminished the HGF/FGF2-induced expression of Sema3A down to a baseline level. Pretreatment with heparitinase and chondroitinase ABC decreased the HGF and FGF2 responses, respectively, in non-knockdown cultures, supporting a possible model that HGF and FGF2 may bind to heparan and chondroitin sulfate chains of syndecan-2, 4 to signal Sema3A expression. The findings, therefore, extend our understanding that HGF/FGF2-syndecan-2, 4 association may stimulate a burst of Sema3A secretion by myoblasts recruited to the site of muscle injury; this would ensure a coordinated delay in the attachment of motoneuron terminals onto fibers early in muscle regeneration, and thus synchronize the recovery of muscle fiber integrity and the early resolution of inflammation after injury with reinnervation toward functional recovery.

7.
PLoS One ; 10(7): e0134303, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26222548

RESUMEN

A recent study demonstrated a positive effect of apple polyphenol (APP) intake on muscle endurance of young-adult animals. While an enhancement of lipid metabolism may be responsible, in part, for the improvement, the contributing mechanisms still need clarification. Here we show that an 8-week intake of 5% (w/w) APP in the diet, up-regulates two features related to fiber type: the ratio of myosin heavy chain (MyHC) type IIx/IIb and myoglobin protein expression in plantaris muscle of 9-week-old male Fischer F344 rats compared to pair-fed controls (P < 0.05). Results were demonstrated by our SDS-PAGE system specialized for MyHC isoform separation and western blotting of whole muscles. Animal-growth profiles (food intake, body-weight gain, and internal-organ weights) did not differ between the control and 5% APP-fed animals (n = 9/group). Findings may account for the increase in fatigue resistance of lower hind limb muscles, as evidenced by a slower decline in the maximum isometric planter-flexion torque generated by a 100-s train of electrical stimulation of the tibial nerve. Additionally, the fatigue resistance was lower after 8 weeks of a 0.5% APP diet than after 5% APP, supporting an APP-dose dependency of the shift in fiber-type composition. Therefore, the present study highlights a promising contribution of dietary APP intake to increasing endurance based on fiber-type composition in rat muscle. Results may help in developing a novel strategy for application in animal sciences, and human sports and age-related health sciences.


Asunto(s)
Malus , Fibras Musculares Esqueléticas/fisiología , Resistencia Física/fisiología , Fitoquímicos/administración & dosificación , Polifenoles/administración & dosificación , Animales , Estimulación Eléctrica , Humanos , Contracción Isométrica/fisiología , Masculino , Músculo Esquelético/fisiología , Mioglobina/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Isoformas de Proteínas/fisiología , Ratas , Ratas Endogámicas F344 , Miosinas del Músculo Esquelético/metabolismo
8.
Anim Sci J ; 85(12): 994-1000, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25185534

RESUMEN

Regenerative intramuscular motor-innervation is thought to reside in the spatiotemporal expression of axon-guidance molecules. Our previous studies showed that resident myogenic stem cells, satellite cells, up-regulate a secreted neural-chemorepellent semaphorin 3A (Sema3A) during the early-differentiation period, in response to hepatocyte growth factor (HGF) elevated in injured muscle. However, a paracrine source of the HGF release is still unknown. Very recently, we proposed a possible contribution of anti-inflammatory macrophages (CD206-positive M2) by showing that M2 cells infiltrate predominantly at the early-differentiation phase (3-5 days post-injury) and produce/secrete large amounts of HGF. However, in understanding this concept there still remains a critical need to examine if phagocytotic pro-inflammatory macrophages (CD86-positive M1), another activated-phenotype still present at the early-differentiation phase concerned, produce HGF upon muscle injury. The current immunocytochemical study demonstrated that the HGF expression is negative for M1 prepared from cardiotoxin-injured Tibialis anterior muscle at day 5, in contrast to the intense fluorescent-signal of M2 served as a positive control. This supplementary result advances our understanding of a spatiotemporal burst of HGF secretion from M2 populations (not M1) to impact Sema3A expression, which ensures a coordinated delay in attachment of motoneuron terminals onto damaged and generating fibers during the early phase of muscle regeneration.


Asunto(s)
Factor de Crecimiento de Hepatocito/análisis , Factor de Crecimiento de Hepatocito/biosíntesis , Activación de Macrófagos/genética , Macrófagos/metabolismo , Músculo Esquelético/fisiología , Regeneración , Animales , Inmunohistoquímica/métodos , Masculino , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Músculo Esquelético/inervación , Semaforina-3A/metabolismo
9.
Int J Biochem Cell Biol ; 54: 272-85, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24886696

RESUMEN

Regenerative mechanisms that regulate intramuscular motor innervation are thought to reside in the spatiotemporal expression of axon-guidance molecules. Our previous studies proposed a heretofore unexplored role of resident myogenic stem cell (satellite cell)-derived myoblasts as a key presenter of a secreted neural chemorepellent semaphorin 3A (Sema3A); hepatocyte growth factor (HGF) triggered its expression exclusively at the early-differentiation phase. In order to verify this concept, the present study was designed to clarify a paracrine source of HGF release. In vitro experiments demonstrated that activated anti-inflammatory macrophages (CD206-positive M2) produce HGF and thereby promote myoblast chemoattraction and Sema3A expression. Media from pro-inflammatory macrophage cultures (M1) did not show any significant effect. M2 also enhanced the expression of myoblast-differentiation markers in culture, and infiltrated predominantly at the early-differentiation phase (3-5 days post-injury); M2 were confirmed to produce HGF as monitored by in vivo/ex vivo immunocytochemistry of CD11b/CD206/HGF-positive cells and by HGF in situ hybridization of cardiotoxin- or crush-injured tibialis anterior muscle, respectively. These studies advance our understanding of the stage-specific activation of Sema3A expression signaling. Findings, therefore, encourage the idea that M2 contribute to spatiotemporal up-regulation of extracellular Sema3A concentrations by producing HGF that, in turn, stimulates a burst of Sema3A secretion by myoblasts that are recruited to site of injury. This model may ensure a coordinated delay in re-attachment of motoneuron terminals onto damaged fibers early in muscle regeneration, and thus synchronize the recovery of muscle-fiber integrity and the early resolution of inflammation after injury.


Asunto(s)
Diferenciación Celular , Movimiento Celular , Macrófagos/inmunología , Neuronas Motoras/citología , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/metabolismo , Regeneración Nerviosa/fisiología , Semaforina-3A/metabolismo , Animales , Antiinflamatorios/metabolismo , Western Blotting , Células Cultivadas , Factor de Crecimiento de Hepatocito/metabolismo , Hibridación in Situ , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/inmunología , Neuronas Motoras/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Mioblastos Esqueléticos/citología , Mioblastos Esqueléticos/efectos de los fármacos , Neurogénesis/fisiología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Semaforina-3A/genética , Transducción de Señal
10.
Anim Sci J ; 84(11): 744-50, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23980916

RESUMEN

Muscle damage induces massive macrophage infiltration of the injury site, in which activated pro-inflammatory and anti-inflammatory phenotypes (currently classified as M1 and M2, respectively) have been documented as distinct functional populations predominant at different times after the conventional acute injury by intramuscular injection of snake venoms (cardiotoxin, notexin) or chemicals (bupivacaine hydrochloride, barium chloride). The present study employed a muscle-crush injury model that may better reflect the physiologic damage and repair processes initiated by contusing a gastrocnemius muscle in the lower hind-limb of adult mice with hemostat forceps, and examined the time-course invasion of M1 and M2 macrophages during muscle regeneration by immunocytochemistry of CD197 and CD206 marker proteins. CD197-positive M1 macrophages were observed exclusively at 1-4 days after crush followed by the alternative prevalence of CD206-positive M2 at 7 days of myogenic differentiation, characterized by increasing levels of myogenin messenger RNA expression. Preliminary PCR analysis showed that M2 may produce hepatocyte growth factor (HGF) in culture, providing additional benefit to understanding that M2 populations actively promote regenerative myogenesis (muscle fiber repair) and moto-neuritogenesis (re-attachment of motoneuron terminals onto damaged fibers) through their time-specific infiltration and release of growth factor at the injury site early in muscle regeneration.


Asunto(s)
Macrófagos/patología , Músculo Esquelético/lesiones , Animales , Factor de Crecimiento de Hepatocito/biosíntesis , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Desarrollo de Músculos/fisiología , Neurogénesis/fisiología , ARN Mensajero/análisis , Factores de Tiempo , Ponzoñas/farmacología
11.
Anim Sci J ; 84(2): 185-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23384361

RESUMEN

Regenerative mechanisms that regulate intramuscular motor innervation. including configuration of the neuromuscular connections are thought to reside in the spatiotemporal expression of axon-guidance molecules. Our previous studies proposed a heretofore unexplored role of satellite cells as a key source of a secreted neural chemorepellent semaphorin 3A (Sema3A) expression. In order to verify this concept, there is still a critical need to provide direct evidence to show the up-regulation of Sema3A protein in satellite cells in vivo upon muscle injury. The present study employed a Sema3A/MyoD double-immunohistochemical staining for cryo-sections prepared from cardiotoxin injected gastrocnemius muscle of adult mouse lower hind-limb. Results clearly demonstrated that Sema3A expression was up-regulated in myogenic differentiation-positive satellite cells at 4-12 days post-injury period, the time that corresponds to the cell differentiation phase characterized by increasing myogenin messenger RNA expression. This direct proof encourages a possible implication of satellite cells in the spatiotemporal regulation of extracellular Sema3A concentrations, which potentially ensures coordinating a delay in neurite sprouting and re-attachment of motoneuron terminals onto damaged muscle fibers early in muscle regeneration in synchrony with recovery of muscle-fiber integrity.


Asunto(s)
Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/fisiología , Semaforina-3A/metabolismo , Animales , Diferenciación Celular , Proteínas Cardiotóxicas de Elápidos/administración & dosificación , Proteínas Cardiotóxicas de Elápidos/toxicidad , Inyecciones Intramusculares , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Regeneración , Células Satélite del Músculo Esquelético/citología , Semaforina-3A/fisiología , Regulación hacia Arriba
12.
Int J Biochem Cell Biol ; 45(2): 476-82, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23085379

RESUMEN

Resident myogenic stem cells, satellite cells, up-regulate a secreted multi-functional modulator, semaphorin 3A (Sema3A), exclusively at the early-differentiation phase in response to muscle-crush injury and treatment with hepatocyte growth factor (HGF) or basic fibroblast growth factor (FGF2). Here, we add evidence that the Sema3A expression and secretion induced by the growth factors is significantly higher in primary cultures from adult rat soleus than from the fast-twitch extensor digitorum longus (EDL) muscle. The higher Sema3A response, revealed by quantitative PCR and Western blotting of cell lysates and conditioned media, may account for the higher myogenin expression of soleus muscle satellite cells early in differentiation since addition of recombinant Sema3A stimulates myogenin expression in cultures. These experiments also showed that mRNA expression of plexin A2, which together with neuropilins, constitutes Sema3A composite-receptors, was higher in satellite cells from soleus than EDL with no difference in plexin A1 and A3 and neuropilin-1 and 2 levels. These comparative studies, therefore, highlight a possible Sema3A-plexin A2-myogenin signaling axis that may ensure promoting early differentiation by soleus muscle satellite cells.


Asunto(s)
Expresión Génica , Músculo Esquelético/citología , Miogenina/genética , Células Satélite del Músculo Esquelético/metabolismo , Semaforina-3A/metabolismo , Animales , Células Cultivadas , Factor 2 de Crecimiento de Fibroblastos/fisiología , Regulación de la Expresión Génica , Factor de Crecimiento de Hepatocito/fisiología , Hipoxantina Fosforribosiltransferasa/genética , Hipoxantina Fosforribosiltransferasa/metabolismo , Masculino , Miogenina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/metabolismo , Estándares de Referencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/normas
13.
Anim Sci J ; 83(10): 712-7, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23035711

RESUMEN

Successful regeneration and remodeling of neuromuscular junctions are critical for restoring functional capacities and properties of skeletal muscle after damage, and axon-guidance molecules may be involved in the signaling that regulates such restoration. Recently, we found that early-differentiated satellite cells up-regulate a secreted neural chemorepellent Sema3A upon in vivo muscle-crush injury. The study also revealed that Sema3A expression is up-regulated in primary satellite-cell cultures in response to hepatocyte growth factor (HGF) and basic fibroblast growth factor (FGF2) and is prevented by transforming growth factor (TGF)-ß2, 3. In order to verify the physiological significance of this regulation in vitro, the present study was designed to estimate the time-course of extracellular HGF, FGF2 and TGF-ß3 concentrations after crush-injury of Gastrocnemius muscle in the rat lower hind-limb, using a combination of a non-homogenization/non-spin extraction of extracellular wound fluids and enhanced chemiluminescence-Western blotting analyses. Results clearly demonstrated that active HGF and FGF2 are prevalent in 2-8 days post-crush, whereas active TGF-ß3 increases after 12 days, providing a better understanding of the time-coordinated levels of HGF, FGF2 and TGF-ß3 that drive regulation of Sema3A expression during regenerative intramuscular moto-neuritogenesis.


Asunto(s)
Factores de Crecimiento de Fibroblastos/análisis , Factor de Crecimiento de Hepatocito/análisis , Músculo Esquelético/lesiones , Factor de Crecimiento Transformador beta3/análisis , Animales , Western Blotting , Ratas , Semaforina-3A/metabolismo , Factores de Tiempo
14.
Am J Physiol Cell Physiol ; 302(12): C1741-50, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22460715

RESUMEN

When skeletal muscle is stretched or injured, satellite cells, resident myogenic stem cells positioned beneath the basal lamina of mature muscle fibers, are activated to enter the cell cycle. This signaling pathway is a cascade of events including calcium-calmodulin formation, nitric oxide (NO) radical production by NO synthase, matrix metalloproteinase activation, release of hepatocyte growth factor (HGF) from the extracellular matrix, and presentation of HGF to the receptor c-met, as demonstrated by assays of primary cultures and in vivo experiments. Here, we add evidence that two ion channels, the mechanosensitive cation channel (MS channel) and the long-lasting-type voltage-gated calcium-ion channel (L-VGC channel), mediate the influx of extracellular calcium ions in response to cyclic stretch in satellite cell cultures. When applied to 1-h stretch cultures with individual inhibitors for MS and L-VGC channels (GsMTx-4 and nifedipine, respectively) or with a less specific inhibitor (gadolinium chloride, Gd), satellite cell activation and upstream HGF release were abolished, as revealed by bromodeoxyuridine-incorporation assays and Western blotting of conditioned media, respectively. The inhibition was dose dependent with a maximum at 0.1 µM (GsMTx-4), 10 µM (nifedipine), or 100 µM (Gd) and canceled by addition of HGF to the culture media; a potent inhibitor for transient-type VGC channels (NNC55-0396, 100 µM) did not show any significant inhibitory effect. The stretch response was also abolished when calcium-chelator EGTA (1.8 mM) was added to the medium, indicating the significance of extracellular free calcium ions in our present activation model. Finally, cation/calcium channel dependencies were further documented by calcium-imaging analyses on stretched cells; results clearly demonstrated that calcium ion influx was abolished by GsMTx-4, nifedipine, and EGTA. Therefore, these results provide an additional insight that calcium ions may flow in through L-VGC channels by possible coupling with adjacent MS channel gating that promotes the local depolarization of cell membranes to initiate the satellite cell activation cascade.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Activación del Canal Iónico , Mecanotransducción Celular , Células Satélite del Músculo Esquelético/metabolismo , Animales , Bencimidazoles/farmacología , Western Blotting , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Quelantes/farmacología , Medios de Cultivo Condicionados/metabolismo , Ciclopropanos/farmacología , Relación Dosis-Respuesta a Droga , Ácido Egtácico/farmacología , Gadolinio/farmacología , Factor de Crecimiento de Hepatocito/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Masculino , Mecanotransducción Celular/efectos de los fármacos , Potenciales de la Membrana , Microscopía Fluorescente , Naftalenos/farmacología , Nifedipino/farmacología , Péptidos/farmacología , Ratas , Células Satélite del Músculo Esquelético/efectos de los fármacos , Venenos de Araña/farmacología , Estrés Mecánico , Factores de Tiempo
15.
Am J Physiol Cell Physiol ; 301(5): C1270-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21865583

RESUMEN

Successful regeneration and remodeling of the intramuscular motoneuron network and neuromuscular connections are critical for restoring skeletal muscle function and physiological properties. The regulatory signals of such coordination remain unclear, although axon-guidance molecules may be involved. Recently, satellite cells, resident myogenic stem cells positioned beneath the basal lamina and at high density at the myoneural junction regions of mature fibers, were shown to upregulate a secreted neural chemorepellent semaphorin 3A (Sema3A) in response to in vivo muscle-crush injury. The initial report on that expression centered on the observation that hepatocyte growth factor (HGF), an essential cue in muscle fiber growth and regeneration, remarkably upregulates Sema3A expression in early differentiated satellite cells in vitro [Tatsumi et al., Am J Physiol Cell Physiol 297: C238-C252, 2009]. Here, we address regulatory effects of basic fibroblast growth factor (FGF2) and transforming growth factor (TGF)-ßs on Sema3A expression in satellite cell cultures. When treated with FGF2, Sema3A message and protein were upregulated as revealed by reverse transcription-polymerase chain reaction and immunochemical studies. Sema3A upregulation by FGF2 was dose dependent with a maximum (8- to 1-fold relative to the control) at 2.5 ng/ml (150 pM) and occurred exclusively at the early differentiation stage. The response was highly comparable in dose response and timing to effects of HGF treatment, without any additive or synergistic effect from treatment with a combination of both potent upregulators. In contrast, TGF-ß2 and -ß3 potently decreased basal Sema3A expression; the maximum effect was at very low concentrations (40 and 8 pM, respectively) and completely cancelled the activities of FGF2 and HGF to upregulate Sema3A. These results therefore encourage the prospect that a time-coordinated increase in HGF, FGF2, and TGF-ß ligands and their receptors promotes a programmed strategy for Sema3A expression that guarantees successful intramuscular motor reinnervation by delaying sprouting and reattachment of motoneuron terminals onto damaged muscle fibers early in regeneration pending restoration of muscle fiber contractile integrity.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Semaforina-3A/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Factor de Crecimiento Transformador beta3/metabolismo , Animales , Células Cultivadas , Factor de Crecimiento de Hepatocito/metabolismo , Masculino , Desarrollo de Músculos , Músculo Esquelético/lesiones , Músculo Esquelético/inervación , Músculo Esquelético/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba , Cicatrización de Heridas
16.
Anim Sci J ; 81(2): 245-51, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20438507

RESUMEN

Satellite cells, resident myogenic stem cells found in postnatal skeletal muscle, are most abundant during early postnatal development and sharply decline in frequency thereafter to adult levels in mice and rats. Therefore, postnatal changes in satellite cell mitotic activities are important aspects for further understanding a muscle growth strategy. In large meat-production animals, however, the traditional in vivo proliferation assay may be less realistic because it requires intra-peritoneal (ip) injection of huge dosage of mutagenic nucleosides, (3)H-labeled thymidine or bromodeoxyuridine (BrdU), at each age-time of sacrifice. We report in the present pilot study using rats that in vivo proliferation activity of satellite cells can be evaluated by an in vitro BrdU-incorporation assay in early cultures. Briefly, satellite cells were prepared from upper hind-limb and back muscles and maintained for 24 h with imposing by BrdU addition for the last 2 h, followed by the regular immunocytochemistry for determining BrdU-incorporated cell percentage. This in vitro assay demonstrated a rapid decrease in proliferating satellite cell frequency to the adult level during about 3-month period after birth, and yielded a high correlation to the measurements by the in vivo BrdU ip-injection method during the postnatal period examined from day-2 to month-11. The in vitro proliferation assay may be further adaptable for large domestic animals by the combination with a muscle biopsy technique that enables age-interval sampling from the same growing animals.


Asunto(s)
Músculo Esquelético/citología , Músculo Esquelético/crecimiento & desarrollo , Células Satélite del Músculo Esquelético/citología , Animales , Bromodesoxiuridina/metabolismo , Proliferación Celular , Técnicas Citológicas , Masculino , Proyectos Piloto , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...