Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Alcohol Clin Exp Res ; 43(7): 1427-1438, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31081931

RESUMEN

BACKGROUND: Alcohol exposure induces TGFß1 and renders the lung susceptible to injury and disrepair. We determined that TGFß1 regulates myofibroblast differentiation through the loss of Thy-1 expression and consequent induction of α-SMA. TGFß1 is important for T helper 17 (Th17) differentiation and IL-17 secretion, which in turn participates in tissue repair. We hypothesized that alcohol induces Th17 differentiation via TGFß1 and that IL-17 produced by these cells contributes to the development of profibrotic lung myofibroblasts. METHODS: Primary lung fibroblasts (PLFs) were treated with alcohol, TGFß1, and IL-17 and then analyzed for Thy-1 expression and cell morphology. Naïve and Th17-polarized CD4+ T cells were exposed to alcohol and assessed for IL-17 expression. CD4+ T cells from alcohol-fed mice were analyzed for Th17 and IL-17 expression. Lungs of control-fed, bleomycin-treated and alcohol-fed, bleomycin-treated mice were analyzed for IL-17 protein expression. RESULTS: Alcohol-treated PLFs expressed lower levels of Thy-1 than untreated cells. TGFß1 or IL-17 exposure suppressed PLF Thy-1 expression. When administered together, TGFß1 and IL-17 additively down-regulated Thy-1 expression. Exposure of naïve and Th17-polarized CD4+ T cells to alcohol induced the Th17 phenotype and augmented their production of IL-17. CD4+ Th17+ levels are elevated in the peripheral compartment but not in the lungs of alcohol-fed animals. Treatment of the PLFs with IL-17 and alcohol induced α-SMA expression. Induction of α-SMA and myofibroblast morphology by IL-17 occurred selectively in a Thy-1- fibroblast subpopulation. Chronic alcohol ingestion augmented lung-specific IL-17 expression following bleomycin-induced lung injury. CONCLUSIONS: Alcohol exposure skews T cells toward a Th17 immune response that in turn primes the lung for fibroproliferative disrepair through loss of Thy-1 expression and induction of myofibroblast differentiation. These effects suggest that IL-17 and TGFß1 contribute to fibroproliferative disrepair in the lung and targeting these proteins could limit morbidity and mortality following lung injury in alcoholic individuals.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Fibroblastos/efectos de los fármacos , Interleucina-17/biosíntesis , Pulmón/patología , Miofibroblastos/efectos de los fármacos , Antígenos Thy-1/biosíntesis , Antígenos Thy-1/genética , Actinas/biosíntesis , Actinas/genética , Animales , Recuento de Linfocito CD4 , Transdiferenciación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Pulmón/efectos de los fármacos , Linfotoxina-alfa/biosíntesis , Ratones , Ratones Endogámicos C57BL , Linfocitos T/efectos de los fármacos
2.
Alcohol Clin Exp Res ; 41(11): 1875-1885, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28888052

RESUMEN

BACKGROUND: We previously demonstrated that chronic alcohol ingestion augments TGFß1 expression in the lung fibroblast and increases the risk of fibroproliferative disrepair in a mouse model of acute lung injury. The effect of alcohol on TGFß1 is mitigated by treatment with sulforaphane (SFP), which can activate nuclear factor (erythroid-derived 2)-like 2 (Nrf2). However, the mechanisms by which alcohol amplifies, or SFP attenuates, TGFß1 expression in the fibroblast are not known. MicroRNA (miR)-21 has been shown to inhibit Smad7, a TGFß1 signaling inhibitor. In this study, we hypothesized that alcohol augments TGFß1 expression through up-regulation of miR-21, which subsequently inhibits Smad7. METHODS: Primary mouse lung fibroblasts were cultured ± alcohol ± SFP and assessed for gene expression of miR-21, and gene and/or protein expression of Nrf2, Nrf2-regulated antioxidant enzymes, Smad7, STAT3, and TGFß1. NIH 3T3 fibroblasts were transfected with a miR-21 inhibitor and cultured ± alcohol. α-SMA, Smad7, and TGFß1 protein expression were then assessed. In parallel, NIH 3T3 lung fibroblasts were transfected with Nrf2 silencing RNA (siRNA) and cultured ± alcohol ± SFP. Gene expression of miR-21, Nrf2, Smad7, and TGFß1 was assessed. RESULTS: MiR-21 gene expression was increased by 12-fold at 48 hours, and Smad7 gene expression and protein expression were reduced by ~30% in alcohol-treated fibroblasts. In parallel, inhibition of miR-21 attenuated alcohol-mediated decrease in Smad7 and increase in TGFß1 and α-SMA protein expression. Treatment with SFP mitigated the effect of alcohol on miR-21, Smad7 and total and phosphorylated STAT3, and restored Nrf2-regulated antioxidant gene expression. Silencing of Nrf2 prevented the effect of SFP on miR-21, Smad7, and TGFß1 gene expression in alcohol-treated NIH 3T3 fibroblasts. CONCLUSIONS: Alcohol treatment increases TGFß1 in fibroblasts, at least in part, through augmentation of miR-21, which then inhibits Smad7 expression. These effects can be attenuated by activation of Nrf2 with SFP.


Asunto(s)
Etanol/farmacología , Fibroblastos/metabolismo , MicroARNs/biosíntesis , Factor 2 Relacionado con NF-E2/biosíntesis , Proteína smad7/biosíntesis , Factor de Crecimiento Transformador beta1/biosíntesis , Animales , Células Cultivadas , Fibroblastos/efectos de los fármacos , Pulmón/citología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Células 3T3 NIH , Proteína smad7/antagonistas & inhibidores
3.
Alcohol Clin Exp Res ; 40(9): 1846-56, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27436123

RESUMEN

BACKGROUND: Alcohol abuse, which impairs antioxidant defenses and promotes acute lung injury, increases Nrf2 nuclear translocation but nevertheless inhibits its activation of the antioxidant response element (ARE). Thioredoxin-1 (Trx1) is required for optimal Nrf2 binding and activation of the ARE, and we hypothesized that its inhibition contributes to impaired Nrf2-ARE signaling in the alcoholic lung. METHODS: Lung tissue and primary lung fibroblasts (PLFs) were isolated from C57/BL6 wild-type (WT) and transgenic mice overexpressing the human Trx1 gene with a nuclear localizing sequence (NLS-Tg); some mice consumed alcohol in water prior to lung tissue and PLF isolation; in some mice, acute lung injury was induced with intratracheal bleomycin. In other experiments, PLFs were isolated from WT and NLS-Tg mice and then exposed to alcohol. Finally, PLF isolated from WT mice were transfected with Trx1 expression vector containing either a cytosolic localized sequence (NES) or a nuclear localized sequence (NLS) prior to alcohol exposure. RESULTS: Alcohol treatment in vivo or in vitro decreased Trx1 expression, and bleomycin-treated alcohol-fed mice had fibrotic disrepair in their lungs. In parallel, whereas alcohol exposure in vitro increased TGFß1 expression and decreased Nrf2-ARE activity in PLF from WT mice, these effects were not observed in PLF from NLS-Tg mice. Finally, selective overexpression of Trx1 in the nucleus but not in the cytosol preserved Nrf2-ARE activity during alcohol exposure. CONCLUSIONS: Although alcohol-induced redox stress actually promotes Nrf2 nuclear translocation, the coincident suppression of Trx1 impairs Nrf2-ARE activity within the nuclear compartment. Nuclear overexpression of Trx1 restored Nrf2-ARE activity and attenuated alcohol-induced TGFß1 expression and alcohol-induced exaggerate response to bleomycin-induced acute lung injury.


Asunto(s)
Núcleo Celular/metabolismo , Etanol/administración & dosificación , Factor 2 Relacionado con NF-E2/metabolismo , Fibrosis Pulmonar/metabolismo , Transducción de Señal/fisiología , Tiorredoxinas/biosíntesis , Animales , Núcleo Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/prevención & control , Transducción de Señal/efectos de los fármacos , Tiorredoxinas/genética
4.
Am J Physiol Cell Physiol ; 309(9): C616-26, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26333597

RESUMEN

Idiopathic pulmonary fibrosis is a progressive lung disease that increases in incidence with age. We identified a profibrotic lung phenotype in aging mice characterized by an increase in the number of fibroblasts lacking the expression of thymocyte differentiation antigen 1 (Thy-1) and an increase in transforming growth factor (TGF)-ß1 expression. It has been shown that Thy-1 expression can be epigenetically modified. Lung fibroblasts (PLFs) were treated with TGF-ß1 ± DNA methyltransferase (DNMT) inhibitor 5-aza-2'-deoxycytidine (5-AZA) and analyzed for Thy-1 gene and protein expression, DNMT protein expression, and activity. α-Smooth muscle actin (α-SMA) and collagen type 1 (Col1A1) gene and protein expression was assessed. PLFs were transfected with DNMT1 silencing RNA ± TGF-ß1. TGF-ß1 inhibited Thy-1 gene and protein expression in PLFs, and cotreatment with 5-AZA ameliorated this effect and appeared to inhibit DNMT1 activation. TGF-ß1 induced Thy-1 promoter methylation as assessed by quantitative methyl PCR. Treatment with 5-AZA attenuated TGF-ß1-induced Col1A1 gene and protein expression and α-SMA gene expression (but not α-SMA protein expression). Inhibiting DNMT1 with silencing RNA attenuated TGF-ß1-induced DNMT activity and its downstream suppression of Thy-1 mRNA and protein expression as well as inhibited α-SMA mRNA and Col1A1 mRNA and protein expression, and showed a decreased trend in Thy-1 promoter methylation. Immunofluorescence for α-SMA suggested that 5-AZA inhibited stress fiber formation. These findings suggest that TGF-ß1 epigenetically regulates lung fibroblast phenotype through methylation of the Thy-1 promoter. Targeted inhibition of DNMT in the right clinical context might prevent fibroblast to myofibroblast transdifferentiation and collagen deposition, which in turn could prevent fibrogenesis in the lung and other organs.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibrosis Pulmonar Idiopática/genética , Pulmón/efectos de los fármacos , Antígenos Thy-1/genética , Factor de Crecimiento Transformador beta1/farmacología , Actinas/genética , Actinas/metabolismo , Animales , Secuencia de Bases , Transdiferenciación Celular , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Inhibidores Enzimáticos/farmacología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Fenotipo , Regiones Promotoras Genéticas , Interferencia de ARN , Antígenos Thy-1/metabolismo , Transfección
5.
Alcohol Clin Exp Res ; 38(11): 2731-42, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25421510

RESUMEN

BACKGROUND: Chronic alcohol ingestion induces the expression of transforming growth factor beta-1(TGFß1), inhibits nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-mediated activation of the antioxidant response element (ARE), depletes alveolar glutathione pools, and potentiates acute lung injury. In this study, we examined the mechanistic relationship between TGFß1 and Nrf2-ARE signaling in the experimental alcoholic lung. METHODS: Wild-type mice were treated ± alcohol in drinking water for 8 weeks and their lungs were assessed for Nrf2 expression. In parallel, mouse lung fibroblasts were cultured ± alcohol and treated ± sulforaphane (SFP; an activator of Nrf2), ±TGFß1, ±TGFß1 neutralizing antibody, and/or ±activin receptor-like kinase 5 inhibitors (to block TGß1 receptor signaling) and then analyzed for the expression of Nrf2, Kelch-like ECH-associated protein 1 (Keap1) and TGFß1, Nrf2-ARE activity, and the expression of the Nrf2-ARE-dependent antioxidants glutathione s-transferase theta 2 (GSTT2) and glutamate-cysteine ligase catalytic subunit (GCLC). Finally, silencing RNA (siRNA) of Nrf2 was then performed prior to alcohol exposure and subsequent analysis of TGFß1 expression. RESULTS: Alcohol treatment in vivo or in vitro decreased Nrf2 expression in murine whole lung and lung fibroblasts, respectively. In parallel, alcohol exposure in vitro decreased Keap1 gene and protein expression in lung fibroblasts. Furthermore, alcohol exposure increased TGFß1 expression but decreased Nrf2-ARE activity and expression of the ARE-dependent genes for GSTT2 and GCLC. These effects of alcohol were prevented by treatment with SFP; in contrast, Nrf2 SiRNA expression exacerbated alcohol-induced TGFß1 expression. Finally, TGFß1 treatment directly suppressed Nrf2-ARE activity whereas blocking TGFß1 signaling attenuated alcohol-induced suppression of Nrf2-ARE activity. CONCLUSIONS: Alcohol-induced oxidative stress is mediated by TGFß1, which suppresses Nrf2-ARE-dependent expression of antioxidant defenses and creates a vicious cycle that feeds back to further increase TGFß1 expression. These effects of alcohol can be mitigated by activation of Nrf2, suggesting a potential therapy in individuals at risk for lung injury due to alcohol abuse.


Asunto(s)
Etanol/toxicidad , Fibroblastos/metabolismo , Pulmón/metabolismo , Factor 2 Relacionado con NF-E2/biosíntesis , Factor de Crecimiento Transformador beta1/fisiología , Animales , Línea Celular Transformada , Fibroblastos/efectos de los fármacos , Humanos , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Células 3T3 NIH , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Factor de Crecimiento Transformador beta1/farmacología
6.
Alcohol Clin Exp Res ; 38(2): 336-43, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24033682

RESUMEN

BACKGROUND: Alcohol abuse increases the risk for acute lung injury (ALI). In both experimental models and in clinical studies, chronic alcohol ingestion causes airway oxidative stress and glutathione depletion and increases the expression of transforming growth factor beta-1 (TGFß1), a potent inducer of fibrosis, in the lung. Therefore, we hypothesized that alcohol ingestion could promote aberrant fibrosis following experimental ALI and that treatment with the glutathione precursor s-adenosylmethionine (SAMe) could mitigate these effects. METHODS: Three-month-old C57BL/6 mice were fed standard chow ± alcohol (20% v/v) in their drinking water for 8 weeks and ±SAMe (4% w/v) during the last 4 weeks. ALI was induced by intratracheal instillation of bleomycin (2.5 units/kg), and lungs were assessed histologically at 7 and 14 days for fibrosis and at 14 days for the expression of extracellular matrix proteins and TGFß1. RESULTS: Alcohol ingestion had no apparent effect on lung inflammation at 7 days, but at 14 days after bleomycin treatment, it increased lung tissue collagen deposition, hydroxyproline content, and the release of activated TGFß1 into the airway. In contrast, SAMe supplementation completely mitigated alcohol-induced priming of these aberrant fibrotic changes through decreased TGFß1 expression in the lung. In parallel, SAMe decreased alcohol-induced TGFß1 and Smad3 mRNA expressions by lung fibroblasts in vitro. CONCLUSIONS: These new experimental findings demonstrate that chronic alcohol ingestion renders the experimental mouse lung susceptible to fibrosis following bleomycin-induced ALI, and that these effects are likely driven by alcohol-mediated oxidative stress and its induction and activation of TGFß1.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Bleomicina/toxicidad , Depresores del Sistema Nervioso Central/toxicidad , Etanol/toxicidad , Fibrosis Pulmonar/inducido químicamente , Actinas/biosíntesis , Animales , Antibióticos Antineoplásicos/antagonistas & inhibidores , Bleomicina/antagonistas & inhibidores , Diferenciación Celular/efectos de los fármacos , Depresores del Sistema Nervioso Central/antagonistas & inhibidores , Dieta , Ensayo de Inmunoadsorción Enzimática , Etanol/antagonistas & inhibidores , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Hidroxiprolina/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Neumonía/patología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/prevención & control , ARN Mensajero/biosíntesis , ARN Mensajero/genética , S-Adenosilmetionina/farmacología , Factor de Crecimiento Transformador beta1/biosíntesis
7.
Sci Rep ; 10(1): 19089, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33154445

RESUMEN

Exaggerated transforming growth factor-beta 1 (TGFß1) expression worsens fibroproliferation following bleomycin-induced lung injury in alcohol-fed mice. MicroRNA (miR)-1946a is predicted to bind to the TGFß1 3' untranslated region (UTR), thereby inhibiting its transcription. We hypothesize that alcohol suppresses miR-1946a and induces TGFß1. Primary murine lung fibroblasts (PLFs) were cultured ± alcohol, miR-1946a mimic or inhibitor, and TGFß1 signaling inhibitors. miR-1946a was analyzed after alcohol treatment in vitro and in vivo. TGFß1 expression and TGFß1 3'UTR-luciferase activity was quantified. We showed that alcohol suppressed miR-1946a in the alcohol-fed mouse lungs and PLFs. MiR-1946a inhibitor increased TGFß1 expression in the fibroblast. MiR-1946a mimic treatment suppressed TGFß1 gene expression and TGFß1 3'UTR activity. Overexpression of miR1946a inhibited alcohol-induced TGFß1 gene and protein expression as well as alcohol-induced TGFß1 and α-smooth muscle actin (SMA) protein expression in PLFs. In conclusion, miR-1946a modulates TGFß1 expression through direct interaction with TGFß1 3'UTR. These findings identify a novel mechanism by which alcohol induces TGFß1 in the lung.


Asunto(s)
Etanol/toxicidad , Pulmón/efectos de los fármacos , Pulmón/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Regiones no Traducidas 3' , Actinas/genética , Actinas/metabolismo , Alcoholismo/genética , Alcoholismo/metabolismo , Alcoholismo/patología , Animales , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Pulmón/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Ratones , Ratones Endogámicos C57BL
8.
Adv Biosci Biotechnol ; 5(1): 19-30, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24596659

RESUMEN

Fibrotic lung diseases increase with age. Previously we determined that senescence increases tissue expression of fibronectin EDA (Fn-EDA) and decreases fibroblast expression of Thy-1, and that fibrocytes contribute to fibrosis following bleomycin-induced lung injury in mice. In this study we hypothesized that fibroblasts lacking Thy-1 expression produce an extracellular matrix that promotes fibrocyte retention and myofibroblast transdifferentiation, thereby promoting fibrogenesis. Young and old mice were treated with bleomycin intratracheally; fibrocytes in the bone marrow, blood, and lungs were quantified, and lung fibroblast Thy-1 expression assessed. Bone marrow-derived fibrocytes were cultured on matrices derived from Thy-1(+) or Thy-1(-) fibroblasts ± the pro-fibrotic cytokine TGFß1. Older mice had more fibrocytes in their bone marrows at baseline and more fibrocytes in their lungs following bleomycin treatment. In parallel, lung fibroblasts in older mice had lower expression of Thy-1 at baseline that increased transiently 7 days after bleomycin treatment but then rapidly waned such that 14 days after bleomycin treatment Thy-1 expression was again markedly lower. Fibrocytes cultured on matrices derived from Thy-1(-) fibroblasts + TGFß1 had increased gene expression for collagen type 1, fibronectin, Fn-EDA, and α-smooth muscle actin. In parallel, whereas the matrices derived from Thy-1(-) fibroblasts stimulated phosphorylation of Akt in cultured fibrocytes, the matrices derived from Thy-1(+) fibroblasts induced apoptosis. These findings suggest that senescence increases fibrocyte recruitment to the lung following injury and that loss of Thy-1 expression by lung fibroblasts promotes fibrocyte retention and myofibroblast trans-differentiation that renders the "aging lung" susceptible to fibrosis.

9.
J Cell Sci ; 120(Pt 1): 149-59, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17164296

RESUMEN

Skeletal muscle regeneration depends on myoblast migration, differentiation and myofiber formation. Isoforms of the nuclear factor of activated T cells (NFAT) family of transcription factors display nonredundant roles in skeletal muscle. NFAT5, a new isoform of NFAT, displays many differences from NFATc1-c4. Here, we examine the role of NFAT5 in myogenesis. NFAT5+/- mice displayed a defect in muscle regeneration with fewer myofibers formed at early times after injury. NFAT5 has a muscle-intrinsic function because inhibition of NFAT5 transcriptional activity caused both a migratory and differentiation defect in cultured myoblasts. We identified Cyr61 as a target of NFAT5 signaling in skeletal muscle cells. Addition of Cyr61 to cells expressing inhibitory forms of NFAT5 rescued the migratory phenotype. These results demonstrate a role for NFAT5 in skeletal muscle cell migration and differentiation. Furthermore, as cell-cell interactions are crucial for myoblast differentiation, these data suggest that myoblast migration and differentiation are coupled and that NFAT5 is a key regulator.


Asunto(s)
Músculo Esquelético/citología , Mioblastos/citología , Mioblastos/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Proteína 61 Rica en Cisteína , Proteínas Inmediatas-Precoces/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Músculo Esquelético/fisiología , Miofibrillas/fisiología , Fenotipo , Regeneración/fisiología , Transducción de Señal/fisiología , Transcripción Genética/fisiología
10.
Am J Physiol Cell Physiol ; 290(6): C1651-9, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16467402

RESUMEN

Loss of muscle mass occurs with disease, injury, aging, and inactivity. Restoration of normal muscle mass depends on myofiber growth, the regulation of which is incompletely understood. Cyclooxygenase (COX)-2 is one of two isoforms of COX that catalyzes the synthesis of prostaglandins, paracrine hormones that regulate diverse physiological and pathophysiological processes. Previously, we demonstrated that the COX-2 pathway regulates early stages of myofiber growth during muscle regeneration. However, whether the COX-2 pathway plays a common role in adult myofiber growth or functions specifically during muscle regeneration is unknown. Therefore, we examined the role of COX-2 during myofiber growth following atrophy in mice. Muscle atrophy was induced by hindlimb suspension (HS) for 2 wk, followed by a reloading period, during which mice were treated with either the COX-2-selective inhibitor SC-236 (6 mg x kg(-1) x day(-1)) or vehicle. COX-2 protein was expressed and SC-236 attenuated myofiber growth during reloading in both soleus and plantaris muscles. Attenuated myofiber growth in the soleus was associated with both decreased myonuclear addition and decreased inflammation, whereas neither of these processes mediated the effects of SC-236 on plantaris growth. In addition, COX-2(-/-) satellite cells exhibited impaired activation/proliferation in vitro, suggesting direct regulation of muscle cell activity by COX-2. Together, these data suggest that the COX-2 pathway plays a common regulatory role during various types of muscle growth via multiple mechanisms.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Músculo Esquelético/fisiología , Atrofia Muscular/enzimología , Regeneración/fisiología , Animales , Inhibidores de la Ciclooxigenasa/farmacología , Femenino , Suspensión Trasera , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Pirazoles/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfonamidas/farmacología
11.
J Cell Physiol ; 209(2): 314-21, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16906571

RESUMEN

CD44 is a transmembrane protein that plays a role in cell-cell interactions and motility in a number of cell types. Cell-cell interactions are critical for myoblast differentiation and fusion but whether CD44 regulates myogenesis is unknown. Here, we show that CD44 plays a functional role in early myogenesis. Analyses of myofiber cross-sectional area, after local injury in mouse tibialis anterior (TA) muscles, revealed that growth was transiently delayed in the absence of CD44. A muscle-intrinsic role for CD44 is suggested as primary myoblasts from CD44(-/-) mice displayed attenuated differentiation and subsequent myotube formation at early times in a differentiation-inducing in vitro environment. Chemotaxis of CD44(-/-) myoblasts toward hepatocyte growth factor (HGF) and basic fibroblast growth factor (bFGF) was totally abrogated, although expression of their respective receptors did not appear to differ from wild-type. Furthermore, motility of CD44(-/-) myoblasts was decreased at early stages of differentiation as determined by time-lapse microscopy. Wild-type myoblasts contained two subpopulations of slow- and fast-migrating cells, whereas CD44(-/-) myoblasts were composed predominantly of the slower migrating subpopulation. Taken together, these data suggest that myoblast migration and differentiation are closely linked and CD44 is a key regulator.


Asunto(s)
Diferenciación Celular/fisiología , Quimiotaxis/fisiología , Receptores de Hialuranos/metabolismo , Mioblastos Esqueléticos/citología , Animales , Diferenciación Celular/efectos de los fármacos , Fusión Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Receptores de Hialuranos/genética , Ratones , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Mioblastos Esqueléticos/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regeneración/efectos de los fármacos
12.
Am J Physiol Cell Physiol ; 282(5): C984-92, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11940513

RESUMEN

Adequate muscle mass is critical for human health. The molecular pathways regulating maintenance and growth of adult skeletal muscle are little understood. Calcineurin (CN) is implicated as a key signaling molecule in hypertrophy. Whether CN is involved in all forms of muscle growth or in different muscles is unknown. Here, we examine the role of CN in regulating maintenance of muscle size and growth of atrophied muscle in the soleus (slow) and plantaris (fast). The CN inhibitor cyclosporin A (CsA) differentially affects muscle growth and maintenance depending on muscle phenotype. The plantaris is more severely affected by CsA than the soleus in both growth conditions. One-week vs. 2-wk CsA treatment suggests that both CN-dependent and CN-independent growth occur in the atrophied soleus, whereas plantaris growth appears to be totally CN dependent. Our results suggest that CN regulates multiple types of muscle growth, depending both on muscle phenotype and stage of myofiber growth. Differential expression of components of the CN pathway occurs and may contribute to the differences between muscles.


Asunto(s)
Calcineurina/metabolismo , Ciclosporina/farmacología , Hipertrofia/fisiopatología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/crecimiento & desarrollo , Animales , Inhibidores de la Calcineurina , Ciclofilinas/metabolismo , Femenino , Suspensión Trasera , Humanos , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Atrofia Muscular/fisiopatología , Isomerasa de Peptidilprolil , Fenotipo , Distribución Aleatoria , Transducción de Señal/fisiología
13.
Am J Physiol Cell Physiol ; 287(2): C475-83, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15084473

RESUMEN

Skeletal muscle regeneration comprises several overlapping cellular processes, including inflammation and myogenesis. Prostaglandins (PGs) may regulate muscle regeneration, because they modulate inflammation and are involved in various stages of myogenesis in vitro. PG synthesis is catalyzed by different isoforms of cyclooxygenase (COX), which are inhibited by nonsteroidal anti-inflammatory drugs. Although experiments employing nonsteroidal anti-inflammatory drugs have implicated PGs in tissue repair, how PGs regulate muscle regeneration remains unclear, and the potentially distinct roles of different COX isoforms have not been investigated. To address these questions, a localized freeze injury was induced in the tibialis anterior muscles of mice chronically treated with either a COX-1- or COX-2-selective inhibitor (SC-560 and SC-236, respectively), starting before injury. The size of regenerating myofibers was analyzed at time points up to 5 wk after injury and found to be decreased by SC-236 and in COX-2(-/-) muscles, but unaffected by SC-560. In contrast, SC-236 had no effect on myofiber growth when administered starting 7 days after injury. The attenuation of myofiber growth by SC-236 treatment and in COX-2(-/-) muscles is associated with decreases in the number of myoblasts and intramuscular inflammatory cells at early times after injury. Together, these data suggest that COX-2-dependent PG synthesis is required during early stages of muscle regeneration and thus raise caution about the use of COX-2-selective inhibitors in patients with muscle injury or disease.


Asunto(s)
Isoenzimas/genética , Isoenzimas/metabolismo , Músculo Esquelético/lesiones , Músculo Esquelético/fisiología , Prostaglandina-Endoperóxido Sintasas/genética , Prostaglandina-Endoperóxido Sintasas/metabolismo , Regeneración/fisiología , Animales , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/farmacología , Regulación Enzimológica de la Expresión Génica , Isoenzimas/antagonistas & inhibidores , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mioblastos/enzimología , Miositis/metabolismo , Miositis/fisiopatología
14.
Cell ; 113(4): 483-94, 2003 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-12757709

RESUMEN

Skeletal muscle formation and growth require the fusion of myoblasts to form multinucleated myofibers or myotubes, but few molecules are known to regulate myoblast fusion in mammals. The transcription factor NFATc2 controls myoblast fusion at a specific stage of myogenesis after the initial formation of a myotube and is necessary for further cell growth. By examining genes regulated by NFATc2 in muscle, this study identifies the cytokine IL-4 as a molecular signal that controls myoblast fusion with myotubes. Muscle cells lacking IL-4 or the IL-4alpha receptor subunit form normally but are reduced in size and myonuclear number. IL-4 is expressed by a subset of muscle cells in fusing muscle cultures and requires the IL-4alpha receptor subunit on myoblasts to promote fusion and growth. These data demonstrate that following myotube formation, myotubes recruit myoblast fusion by secretion of IL-4, leading to muscle growth.


Asunto(s)
Diferenciación Celular/fisiología , Fusión Celular , Interleucina-4/deficiencia , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/crecimiento & desarrollo , Mioblastos/metabolismo , Proteínas Nucleares , Animales , Tamaño de la Célula/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Interleucina-4/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos/citología , Factores de Transcripción NFATC , Receptores de Interleucina-4/deficiencia , Receptores de Interleucina-4/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA