Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Clin Sci (Lond) ; 129(12): 1237-49, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26415649

RESUMO

The cytokine transforming growth factor (TGF)-ß1 plays a central role in diabetic nephropathy (DN) with data implicating the miRNA (miR) miR-21 as a key modulator of its prosclerotic actions. In the present study, we demonstrate data indicating that miR-21 up-regulation positively correlates with the severity of fibrosis and rate of decline in renal function in human DN. Furthermore, concomitant analyses of various models of fibrotic renal disease and experimental DN, confirm tubular miR-21 up-regulation. The fibrotic changes associated with increased miR-21 levels are proposed to include the regulation of TGF-ß1-mediated mothers against decapentaplegic homolog 3 (SMAD3)- and phosphoinositide 3-kinase (PI3K)-dependent signalling pathways via co-ordinated repression of mothers against decapentaplegic homolog 7 (SMAD7) and phosphatase and tensin homologue (PTEN) respectively. This represents a previously uncharacterized interaction axis between miR-21 and PTEN-SMAD7. Targeting of these proteins by miR-21 resulted in de-repression of the respective pathways as reflected by increases in SMAD3 and V-Akt murine thymoma viral oncogene homolog 1 (AKT) phosphorylation. Many of the changes typically induced by TGF-ß1, including phosphorylation of signalling mediators, were further enhanced by miR-21. Collectively, these data present a unified model for a key role for miR-21 in the regulation of renal tubular extracellular matrix (ECM) synthesis and accumulation and provide important insights into the molecular pathways implicated in the progression of DN.


Assuntos
Nefropatias Diabéticas/metabolismo , Túbulos Renais Proximais/metabolismo , MicroRNAs/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteína Smad7/metabolismo , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Estudos de Casos e Controles , Linhagem Celular , Colágeno/metabolismo , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Fibrose , Taxa de Filtração Glomerular , Humanos , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/patologia , Túbulos Renais Proximais/fisiopatologia , Camundongos Knockout , MicroRNAs/genética , Fosforilação , Prognóstico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Ratos Sprague-Dawley , Índice de Gravidade de Doença , Transdução de Sinais , Proteína Smad7/genética , Transfecção , Fator de Crescimento Transformador beta1/farmacologia , Regulação para Cima
2.
FASEB J ; 28(4): 1711-23, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24378873

RESUMO

In models of cancer cachexia, inhibiting type IIB activin receptors (ActRIIBs) reverse muscle wasting and prolongs survival, even with continued tumor growth. ActRIIB mediates signaling of numerous TGF-ß proteins; of these, we demonstrate that activins are the most potent negative regulators of muscle mass. To determine whether activin signaling in the absence of tumor-derived factors induces cachexia, we used recombinant serotype 6 adeno-associated virus (rAAV6) vectors to increase circulating activin A levels in C57BL/6 mice. While mice injected with control vector gained ~10% of their starting body mass (3.8±0.4 g) over 10 wk, mice injected with increasing doses of rAAV6:activin A exhibited weight loss in a dose-dependent manner, to a maximum of -12.4% (-4.2±1.1 g). These reductions in body mass in rAAV6:activin-injected mice correlated inversely with elevated serum activin A levels (7- to 24-fold). Mechanistically, we show that activin A reduces muscle mass and function by stimulating the ActRIIB pathway, leading to deleterious consequences, including increased transcription of atrophy-related ubiquitin ligases, decreased Akt/mTOR-mediated protein synthesis, and a profibrotic response. Critically, we demonstrate that the muscle wasting and fibrosis that ensues in response to excessive activin levels is fully reversible. These findings highlight the therapeutic potential of targeting activins in cachexia.


Assuntos
Ativinas/genética , Caquexia/genética , Expressão Gênica , Atrofia Muscular/genética , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Ativinas/sangue , Ativinas/metabolismo , Animais , Western Blotting , Caquexia/metabolismo , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dependovirus/genética , Vetores Genéticos/genética , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/metabolismo , Miostatina/deficiência , Miostatina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Transdução de Sinais/genética
3.
FASEB J ; 28(12): 5097-110, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25145628

RESUMO

Expression of microRNA-652 (miR-652) increases in the diseased heart, decreases in a setting of cardioprotection, and is inversely correlated with heart function. The aim of this study was to assess the therapeutic potential of inhibiting miR-652 in a mouse model with established pathological hypertrophy and cardiac dysfunction due to pressure overload. Mice were subjected to a sham operation or transverse aortic constriction (TAC) for 4 wk to induce hypertrophy and cardiac dysfunction, followed by administration of a locked nucleic acid (LNA)-antimiR-652 (miR-652 inhibitor) or LNA control. Cardiac function was assessed before and 8 wk post-treatment. Expression of miR-652 increased in hearts subjected to TAC compared to sham surgery (2.9-fold), and this was suppressed by ∼95% in LNA-antimiR-652-treated TAC mice. Inhibition of miR-652 improved cardiac function in TAC mice (fractional shortening:29±1% at 4 wk post-TAC compared to 35±1% post-treatment) and attenuated cardiac hypertrophy. Improvement in heart function was associated with reduced cardiac fibrosis, less apoptosis and B-type natriuretic peptide gene expression, and preserved angiogenesis. Mechanistically, we identified Jagged1 (a Notch1 ligand) as a novel direct target of miR-652. In summary, these studies provide the first evidence that silencing of miR-652 protects the heart against pathological remodeling and improves heart function.


Assuntos
Cardiomegalia/genética , Inativação Gênica , Coração/fisiopatologia , MicroRNAs/genética , Animais , Células Cultivadas , Camundongos , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real
4.
Proc Natl Acad Sci U S A ; 109(43): 17615-20, 2012 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23047694

RESUMO

MicroRNAs are dysregulated in a setting of heart disease and have emerged as promising therapeutic targets. MicroRNA-34 family members (miR-34a, -34b, and -34c) are up-regulated in the heart in response to stress. In this study, we assessed whether inhibition of the miR-34 family using an s.c.-delivered seed-targeting 8-mer locked nucleic acid (LNA)-modified antimiR (LNA-antimiR-34) can provide therapeutic benefit in mice with preexisting pathological cardiac remodeling and dysfunction due to myocardial infarction (MI) or pressure overload via transverse aortic constriction (TAC). An additional cohort of mice subjected to MI was given LNA-antimiR-34a (15-mer) to inhibit miR-34a alone as a comparison for LNA-antimiR-34. LNA-antimiR-34 (8-mer) efficiently silenced all three miR-34 family members in both cardiac stress models and attenuated cardiac remodeling and atrial enlargement. In contrast, inhibition of miR-34a alone with LNA-antimiR-34a (15-mer) provided no benefit in the MI model. In mice subjected to pressure overload, LNA-antimiR-34 improved systolic function and attenuated lung congestion, associated with reduced cardiac fibrosis, increased angiogenesis, increased Akt activity, decreased atrial natriuretic peptide gene expression, and maintenance of sarcoplasmic reticulum Ca(2+) ATPase gene expression. Improved outcome in LNA-antimiR-34-treated MI and TAC mice was accompanied by up-regulation of several direct miR-34 targets, including vascular endothelial growth factors, vinculin, protein O-fucosyltranferase 1, Notch1, and semaphorin 4B. Our results provide evidence that silencing of the entire miR-34 family can protect the heart against pathological cardiac remodeling and improve function. Furthermore, these data underscore the utility of seed-targeting 8-mer LNA-antimiRs in the development of new therapeutic approaches for pharmacologic inhibition of disease-implicated miRNA seed families.


Assuntos
Testes de Função Cardíaca , MicroRNAs/antagonistas & inibidores , Remodelação Ventricular , Animais , Sequência de Bases , DNA , Proteínas de Ligação a DNA/metabolismo , Fucosiltransferases/metabolismo , Camundongos , Dados de Sequência Molecular , Neovascularização Patológica , Oligonucleotídeos/química , Proteínas Proto-Oncogênicas c-bcl-6 , Semaforinas/metabolismo , Regulação para Cima , Vinculina/metabolismo
5.
Clin Exp Pharmacol Physiol ; 41(9): 727-37, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25115402

RESUMO

The identification of non-coding RNA species, previously thought of as 'junk' DNA, adds a new dimension of complexity to the regulation of DNA, RNA and protein. MicroRNAs are short non-coding RNA species that control gene expression, are dysregulated in settings of cardiac and skeletal muscle disease and have emerged as promising therapeutic targets. MicroRNAs specifically enriched in cardiac and skeletal muscle are called myomiRs and play an important role in cardiac pathology and skeletal muscle biology. Moreover, microRNA profiles are altered in response to exercise and disease; thus, their potential as therapeutic drug targets is being widely explored. In the cardiovascular field, therapeutic inhibition of microRNAs has been shown to be effective in improving cardiac outcome in preclinical cardiac disease models. MicroRNAs that promote skeletal muscle regeneration are attractive therapeutic targets in muscle wasting conditions where regenerative capacity is compromised.


Assuntos
Saúde , Cardiopatias/tratamento farmacológico , MicroRNAs/genética , MicroRNAs/metabolismo , Terapia de Alvo Molecular/métodos , Músculo Esquelético/metabolismo , Doenças Musculares/tratamento farmacológico , Miocárdio/metabolismo , Animais , Exercício Físico/fisiologia , Coração/efeitos dos fármacos , Coração/crescimento & desenvolvimento , Cardiopatias/genética , Cardiopatias/metabolismo , Humanos , Músculo Esquelético/crescimento & desenvolvimento , Doenças Musculares/genética , Doenças Musculares/metabolismo
6.
PLoS Genet ; 6(1): e1000816, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20084116

RESUMO

Contributions of null and hypomorphic alleles of Apc in mice produce both developmental and pathophysiological phenotypes. To ascribe the resulting genotype-to-phenotype relationship unambiguously to the Wnt/beta-catenin pathway, we challenged the allele combinations by genetically restricting intracellular beta-catenin expression in the corresponding compound mutant mice. Subsequent evaluation of the extent of resulting Tcf4-reporter activity in mouse embryo fibroblasts enabled genetic measurement of Wnt/beta-catenin signaling in the form of an allelic series of mouse mutants. Different permissive Wnt signaling thresholds appear to be required for the embryonic development of head structures, adult intestinal polyposis, hepatocellular carcinomas, liver zonation, and the development of natural killer cells. Furthermore, we identify a homozygous Apc allele combination with Wnt/beta-catenin signaling capacity similar to that in the germline of the Apc(min) mice, where somatic Apc loss-of-heterozygosity triggers intestinal polyposis, to distinguish whether co-morbidities in Apc(min) mice arise independently of intestinal tumorigenesis. Together, the present genotype-phenotype analysis suggests tissue-specific response levels for the Wnt/beta-catenin pathway that regulate both physiological and pathophysiological conditions.


Assuntos
Camundongos/genética , Camundongos/metabolismo , Transdução de Sinais , beta Catenina/metabolismo , Proteína da Polipose Adenomatosa do Colo/genética , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Células Cultivadas , Embrião de Mamíferos , Feminino , Fibroblastos/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/embriologia , Intestinos/crescimento & desenvolvimento , Fígado/embriologia , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Masculino , Camundongos/embriologia , Camundongos/crescimento & desenvolvimento , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Wnt , Proteína Wnt3 , beta Catenina/genética
7.
J Am Soc Nephrol ; 23(2): 252-65, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22095944

RESUMO

Synthesis and deposition of extracellular matrix (ECM) within the glomerulus and interstitium characterizes renal fibrosis, but the mechanisms underlying this process are incompletely understood. The profibrotic cytokine TGF-ß1 modulates the expression of certain microRNAs (miRNAs), suggesting that miRNAs may have a role in the pathogenesis of renal fibrosis. Here, we exposed proximal tubular cells, primary mesangial cells, and podocytes to TGF-ß1 to examine its effect on miRNAs and subsequent collagen synthesis. TGF-ß1 reduced expression of the miR-29a/b/c/family, which targets collagen gene expression, and increased expression of ECM proteins. In both resting and TGF-ß1-treated cells, ectopic expression of miR-29 repressed the expression of collagens I and IV at both the mRNA and protein levels by targeting the 3'untranslated region of these genes. Furthermore, we observed low levels of miR-29 in three models of renal fibrosis representing early and advanced stages of disease. Administration of the Rho-associated kinase inhibitor fasudil prevented renal fibrosis and restored expression of miR-29. Taken together, these data suggest that TGF-ß1 inhibits expression of the miR-29 family, thereby promoting expression of ECM components. Pharmacologic modulation of these miRNAs may have therapeutic potential for progressive renal fibrosis.


Assuntos
Colágeno/genética , Rim/patologia , MicroRNAs/fisiologia , Regiões 3' não Traduzidas/genética , Animais , Caderinas/genética , Células Cultivadas , Nefropatias Diabéticas/metabolismo , Fibrose , Regulação da Expressão Gênica , Rim/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta1/farmacologia
8.
J Biol Chem ; 286(16): 13805-14, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21324893

RESUMO

MicroRNAs (miRs) are emerging as prominent players in the regulation of many biological processes, including myogenic commitment and skeletal muscle formation. Members of the TGF-ß family can influence the proliferation and myogenic differentiation of cells, although it is presently not clear what role miRNAs play in the TGF-ß-mediated control of myogenic differentiation. Here, we demonstrate in the myogenic C2C12 cell line, and in primary muscle cells, that miR-206 and miR-29-two miRs that act on transcriptional events implicated in muscle differentiation are down-regulated by TGF-ß. We further demonstrate that TGF-ß treatment of myogenic cells is associated with increased expression of histone deacetylase 4 (HDAC4), a key inhibitor of muscle differentiation that has been identified as a target for regulation by miR-206 and miR-29. We confirmed that increased expression of miR-206 and miR-29 resulted in the translational repression of HDAC4 in the presence or absence of TGF-ß via interaction with the HDAC4 3'-untranslated region. Importantly, we found that miR-206 and miR-29 can attenuate the inhibitory actions of TGF-ß on myogenic differentiation. Furthermore, we present evidence that the mechanism by which miR-206 and miR-29 can inhibit the TGF-ß-mediated up-regulation of HDAC4 is via the inhibition of Smad3 expression, a transducer of TGF-ß signaling. These findings identify a novel mechanism of interaction between TGF-ß and miR-206 and -29 in the regulation of myogenic differentiation through HDAC4.


Assuntos
Histona Desacetilases/metabolismo , MicroRNAs/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Regiões 3' não Traduzidas , Animais , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Camundongos , Desenvolvimento Muscular , Músculos/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
9.
J Biomed Biotechnol ; 2011: 212819, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22162630

RESUMO

Recognised by their de novo expression of alpha-smooth muscle actin (SMA), recruitment of myofibroblasts is key to the pathogenesis of fibrosis in chronic kidney disease. Increasingly, we realise that epithelial-mesenchymal transition (EMT) may be an important source of these cells. In this study we describe a novel model of renal EMT. Rat kidney explants were finely diced on gelatin-coated Petri dishes and cultured in serum-supplemented media. Morphology and immunocytochemistry were used to identify mesenchymal (vimentin+, α-smooth muscle actin (SMA)+, desmin+), epithelial (cytokeratin+), and endothelial (RECA+) cells at various time points. Cell outgrowths were all epithelial in origin (cytokeratin+) at day 3. By day 10, 50 ± 12% (mean ± SE) of cytokeratin+ cells double-labelled for SMA, indicating EMT. Lectin staining established a proximal tubule origin. By day 17, cultures consisted only of myofibroblasts (SMA+/cytokeratin-). Explanting is a reproducible ex vivo model of EMT. The ability to modify this change in phenotype provides a useful tool to study the regulation and mechanisms of renal tubulointerstitial fibrosis.


Assuntos
Células Epiteliais/citologia , Transição Epitelial-Mesenquimal/fisiologia , Túbulos Renais Proximais/metabolismo , Rim/citologia , Células-Tronco Mesenquimais/citologia , Miofibroblastos/citologia , Animais , Biomarcadores/análise , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Proliferação de Células , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Imunoquímica/métodos , Células-Tronco Mesenquimais/metabolismo , Miofibroblastos/metabolismo , Fenótipo , Ratos , Ratos Sprague-Dawley , Coloração e Rotulagem/métodos
10.
Pharmaceuticals (Basel) ; 14(3)2021 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-33804032

RESUMO

Cardiac fibrosis is associated with most forms of cardiovascular disease. No reliable therapies targeting cardiac fibrosis are available, thus identifying novel drugs that can resolve or prevent fibrosis is needed. Tilorone, an antiviral agent, can prevent fibrosis in a mouse model of lung disease. We investigated the anti-fibrotic effects of tilorone in human cardiac fibroblasts in vitro by performing a radioisotopic assay for [3H]-proline incorporation as a proxy for collagen synthesis. Exploratory studies in human cardiac fibroblasts treated with tilorone (10 µM) showed a significant reduction in transforming growth factor-ß induced collagen synthesis compared to untreated fibroblasts. To determine if this finding could be recapitulated in vivo, mice with established pathological remodelling due to four weeks of transverse aortic constriction (TAC) were administered tilorone (50 mg/kg, i.p) or saline every third day for eight weeks. Treatment with tilorone was associated with attenuation of fibrosis (assessed by Masson's trichrome stain), a favourable cardiac gene expression profile and no further deterioration of cardiac systolic function determined by echocardiography compared to saline treated TAC mice. These data demonstrate that tilorone has anti-fibrotic actions in human cardiac fibroblasts and the adult mouse heart, and represents a potential novel therapy to treat fibrosis associated with heart failure.

11.
Sci Transl Med ; 13(605)2021 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-34349036

RESUMO

Most patients with advanced solid cancers exhibit features of cachexia, a debilitating syndrome characterized by progressive loss of skeletal muscle mass and strength. Because the underlying mechanisms of this multifactorial syndrome are incompletely defined, effective therapeutics have yet to be developed. Here, we show that diminished bone morphogenetic protein (BMP) signaling is observed early in the onset of skeletal muscle wasting associated with cancer cachexia in mouse models and in patients with cancer. Cancer-mediated factors including Activin A and IL-6 trigger the expression of the BMP inhibitor Noggin in muscle, which blocks the actions of BMPs on muscle fibers and motor nerves, subsequently causing disruption of the neuromuscular junction (NMJ), denervation, and muscle wasting. Increasing BMP signaling in the muscles of tumor-bearing mice by gene delivery or pharmacological means can prevent muscle wasting and preserve measures of NMJ function. The data identify perturbed BMP signaling and denervation of muscle fibers as important pathogenic mechanisms of muscle wasting associated with tumor growth. Collectively, these findings present interventions that promote BMP-mediated signaling as an attractive strategy to counteract the loss of functional musculature in patients with cancer.


Assuntos
Caquexia , Neoplasias , Animais , Denervação , Humanos , Camundongos , Músculo Esquelético/patologia , Atrofia Muscular , Neoplasias/complicações , Neoplasias/patologia
12.
Int J Biochem Cell Biol ; 39(1): 206-19, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-16973406

RESUMO

Tubulointerstitial fibrosis is largely mediated by (myo)fibroblasts present in the interstitium. In this study, we investigated the role of mTOR and phosphatidylinositol 3-kinase in the regulation of fibroblast kinetics, fibroblast differentiation, and collagen synthesis. Rat renal fibroblasts were propagated from kidneys 3 days post-ureteric obstruction and specific inhibitors of mTOR (RAD) and phosphatidylinositol 3-kinase (LY294002) were used to examine the regulation of fibrogenesis. LY294002 but not RAD completely inhibited phosphorylation of Akt, while both inhibitors decreased phosphorylation of the S6 ribosomal protein. RAD and LY decreased foetal calf serum stimulated proliferation and DNA synthesis. In addition to their individual effects, treatment with both RAD and LY294002 decreased serum-induced fibroblast proliferation and DNA synthesis significantly more than either drug alone. TUNEL positive cells (apoptosis) in RAD and LY294002 treated groups were not different from control groups. In addition to their effect on proliferation, both inhibitors also reduced total collagen synthesis. Differentiation studies indicated an increase in alpha-smooth muscle actin expression relative to beta-actin (western blotting), with cytochemistry confirming that all doses of RAD and LY294002 increased the proportion of alpha-smooth muscle actin positive cells, and hence myofibroblasts. Effects were independent of cell toxicity. These results highlight the potential significance of PI3K and mTOR, in the regulation of renal (myo)fibroblast activity. The synergistic effects of LY and RAD on proliferation suggest that mTOR signalling involves pathways other than phosphatidylinositol 3-kinase. These results provide a novel insight into the mechanisms of fibroblast regulation during fibrogenesis.


Assuntos
Diferenciação Celular/fisiologia , Fibroblastos/enzimologia , Rim/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais/fisiologia , Actinas/biossíntese , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Fibroblastos/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Rim/citologia , Morfolinas/farmacologia , Células Musculares/citologia , Células Musculares/enzimologia , Inibidores de Fosfoinositídeo-3 Quinase , Ratos , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR
13.
Sci Transl Med ; 8(348): 348ra98, 2016 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-27440729

RESUMO

Patients with advanced cancer often succumb to complications arising from striated muscle wasting associated with cachexia. Excessive activation of the type IIB activin receptor (ActRIIB) is considered an important mechanism underlying this wasting, where circulating procachectic factors bind ActRIIB and ultimately lead to the phosphorylation of SMAD2/3. Therapeutics that antagonize the binding of ActRIIB ligands are in clinical development, but concerns exist about achieving efficacy without off-target effects. To protect striated muscle from harmful ActRIIB signaling, and to reduce the risk of off-target effects, we developed an intervention using recombinant adeno-associated viral vectors (rAAV vectors) that increase expression of Smad7 in skeletal and cardiac muscles. SMAD7 acts as an intracellular negative regulator that prevents SMAD2/3 activation and promotes degradation of ActRIIB complexes. In mouse models of cachexia, rAAV:Smad7 prevented wasting of skeletal muscles and the heart independent of tumor burden and serum levels of procachectic ligands. Mechanistically, rAAV:Smad7 administration abolished SMAD2/3 signaling downstream of ActRIIB and inhibited expression of the atrophy-related ubiquitin ligases MuRF1 and MAFbx. These findings identify muscle-directed Smad7 gene delivery as a potential approach for preventing muscle wasting under conditions where excessive ActRIIB signaling occurs, such as cancer cachexia.


Assuntos
Atrofia Muscular/metabolismo , Atrofia Muscular/terapia , Neoplasias/fisiopatologia , Proteína Smad7/metabolismo , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Animais , Western Blotting , Camundongos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/etiologia , Miocárdio/metabolismo , Miocárdio/patologia , Neoplasias/complicações , Neoplasias/metabolismo , Fosforilação/genética , Fosforilação/fisiologia , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Proteína Smad7/genética , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
14.
Sci Rep ; 5: 17535, 2015 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-26657343

RESUMO

Follistatin is an inhibitor of TGF-ß superfamily ligands that repress skeletal muscle growth and promote muscle wasting. Accordingly, follistatin has emerged as a potential therapeutic to ameliorate the deleterious effects of muscle atrophy. However, it remains unclear whether the anabolic effects of follistatin are conserved across different modes of non-degenerative muscle wasting. In this study, the delivery of a recombinant adeno-associated viral vector expressing follistatin (rAAV:Fst) to the hind-limb musculature of mice two weeks prior to denervation or tenotomy promoted muscle hypertrophy that was sufficient to preserve muscle mass comparable to that of untreated sham-operated muscles. However, administration of rAAV:Fst to muscles at the time of denervation or tenotomy did not prevent subsequent muscle wasting. Administration of rAAV:Fst to innervated or denervated muscles increased protein synthesis, but markedly reduced protein degradation only in innervated muscles. Phosphorylation of the signalling proteins mTOR and S6RP, which are associated with protein synthesis, was increased in innervated muscles administered rAAV:Fst, but not in treated denervated muscles. These results demonstrate that the anabolic effects of follistatin are influenced by the interaction between muscle fibres and motor nerves. These findings have important implications for understanding the potential efficacy of follistatin-based therapies for non-degenerative muscle wasting.


Assuntos
Denervação/efeitos adversos , Folistatina/genética , Terapia Genética , Atrofia Muscular/etiologia , Atrofia Muscular/patologia , Tenotomia/efeitos adversos , Animais , Dependovirus/genética , Modelos Animais de Doenças , Folistatina/metabolismo , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Hipertrofia , Camundongos , Músculo Esquelético/inervação , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/terapia , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Transdução Genética
15.
PLoS One ; 9(2): e90337, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24587330

RESUMO

Therapeutic inhibition of the miR-34 family (miR-34a,-b,-c), or miR-34a alone, have emerged as promising strategies for the treatment of cardiac pathology. However, before advancing these approaches further for potential entry into the clinic, a more comprehensive assessment of the therapeutic potential of inhibiting miR-34a is required for two key reasons. First, miR-34a has ∼40% fewer predicted targets than the miR-34 family. Hence, in cardiac stress settings in which inhibition of miR-34a provides adequate protection, this approach is likely to result in less potential off-target effects. Secondly, silencing of miR-34a alone may be insufficient in settings of established cardiac pathology. We recently demonstrated that inhibition of the miR-34 family, but not miR-34a alone, provided benefit in a chronic model of myocardial infarction. Inhibition of miR-34 also attenuated cardiac remodeling and improved heart function following pressure overload, however, silencing of miR-34a alone was not examined. The aim of this study was to assess whether inhibition of miR-34a could attenuate cardiac remodeling in a mouse model with pre-existing pathological hypertrophy. Mice were subjected to pressure overload via constriction of the transverse aorta for four weeks and echocardiography was performed to confirm left ventricular hypertrophy and systolic dysfunction. After four weeks of pressure overload (before treatment), two distinct groups of animals became apparent: (1) mice with moderate pathology (fractional shortening decreased ∼20%) and (2) mice with severe pathology (fractional shortening decreased ∼37%). Mice were administered locked nucleic acid (LNA)-antimiR-34a or LNA-control with an eight week follow-up. Inhibition of miR-34a in mice with moderate cardiac pathology attenuated atrial enlargement and maintained cardiac function, but had no significant effect on fetal gene expression or cardiac fibrosis. Inhibition of miR-34a in mice with severe pathology provided no therapeutic benefit. Thus, therapies that inhibit miR-34a alone may have limited potential in settings of established cardiac pathology.


Assuntos
Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/fisiopatologia , Inativação Gênica , MicroRNAs/genética , Animais , Cardiomiopatia Hipertrófica/patologia , Modelos Animais de Doenças , Ecocardiografia , Fibrose , Regulação da Expressão Gênica , Masculino , Camundongos , Índice de Gravidade de Doença , Remodelação Ventricular/genética
16.
PLoS One ; 8(9): e73589, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24023888

RESUMO

microRNAs regulate the development of myogenic progenitors, and the formation of skeletal muscle fibers. However, the role miRNAs play in controlling the growth and adaptation of post-mitotic musculature is less clear. Here, we show that inhibition of the established pro-myogenic regulator miR-206 can promote hypertrophy and increased protein synthesis in post-mitotic cells of the myogenic lineage. We have previously demonstrated that histone deacetylase 4 (HDAC4) is a target of miR-206 in the regulation of myogenic differentiation. We confirmed that inhibition of miR-206 de-repressed HDAC4 accumulation in cultured myotubes. Importantly, inhibition of HDAC4 activity by valproic acid or sodium butyrate prevented hypertrophy of myogenic cells otherwise induced by inhibition of miR-206. To test the significance of miRNA-206 as a regulator of skeletal muscle mass in vivo, we designed recombinant adeno-associated viral vectors (rAAV6 vectors) expressing miR-206, or a miR-206 "sponge," featuring repeats of a validated miR-206 target sequence. We observed that over-expression or inhibition of miR-206 in the muscles of mice decreased or increased endogenous HDAC4 levels respectively, but did not alter muscle mass or myofiber size. We subsequently manipulated miR-206 levels in muscles undergoing follistatin-induced hypertrophy or denervation-induced atrophy (models of muscle adaptation where endogenous miR-206 expression is altered). Vector-mediated manipulation of miR-206 activity in these models of cell growth and wasting did not alter gain or loss of muscle mass respectively. Our data demonstrate that although the miR-206/HDAC4 axis operates in skeletal muscle, the post-natal expression of miR-206 is not a key regulator of basal skeletal muscle mass or specific modes of muscle growth and wasting. These studies support a context-dependent role of miR-206 in regulating hypertrophy that may be dispensable for maintaining or modifying the adult skeletal muscle phenotype--an important consideration in relation to the development of therapeutics designed to manipulate microRNA activity in musculature.


Assuntos
Histona Desacetilases/genética , MicroRNAs/genética , Desenvolvimento Muscular , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/patologia , Animais , Linhagem Celular , Dependovirus/genética , Vetores Genéticos/genética , Hipertrofia/genética , Camundongos , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/fisiologia , Atrofia Muscular/genética
17.
J Cell Biol ; 203(2): 345-57, 2013 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-24145169

RESUMO

Although the canonical transforming growth factor ß signaling pathway represses skeletal muscle growth and promotes muscle wasting, a role in muscle for the parallel bone morphogenetic protein (BMP) signaling pathway has not been defined. We report, for the first time, that the BMP pathway is a positive regulator of muscle mass. Increasing the expression of BMP7 or the activity of BMP receptors in muscles induced hypertrophy that was dependent on Smad1/5-mediated activation of mTOR signaling. In agreement, we observed that BMP signaling is augmented in models of muscle growth. Importantly, stimulation of BMP signaling is essential for conservation of muscle mass after disruption of the neuromuscular junction. Inhibiting the phosphorylation of Smad1/5 exacerbated denervation-induced muscle atrophy via an HDAC4-myogenin-dependent process, whereas increased BMP-Smad1/5 activity protected muscles from denervation-induced wasting. Our studies highlight a novel role for the BMP signaling pathway in promoting muscle growth and inhibiting muscle wasting, which may have significant implications for the development of therapeutics for neuromuscular disorders.


Assuntos
Proteína Morfogenética Óssea 7/metabolismo , Desenvolvimento Muscular , Músculo Esquelético/metabolismo , Atrofia Muscular/prevenção & controle , Transdução de Sinais , Animais , Proteína Morfogenética Óssea 7/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Dependovirus , Modelos Animais de Doenças , Feminino , Folistatina/metabolismo , Terapia Genética , Vetores Genéticos , Células HEK293 , Histona Desacetilases/metabolismo , Humanos , Hipertrofia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/inervação , Músculo Esquelético/patologia , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Miogenina/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Proteínas Smad/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Transdução Genética , Transfecção , Ubiquitina-Proteína Ligases/metabolismo
18.
PLoS One ; 7(12): e51627, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251598

RESUMO

Recombinant adeno-associated viral vectors (rAAV vectors) are promising tools for delivering transgenes to skeletal muscle, in order to study the mechanisms that control the muscle phenotype, and to ameliorate diseases that perturb muscle homeostasis. Many studies have employed rAAV vectors carrying reporter genes encoding for ß-galactosidase (ß-gal), human placental alkaline phosphatase (hPLAP), and green fluorescent protein (GFP) as experimental controls when studying the effects of manipulating other genes. However, it is not clear to what extent these reporter genes can influence signaling and gene expression signatures in skeletal muscle, which may confound the interpretation of results obtained in experimentally manipulated muscles. Herein, we report a strong pro-inflammatory effect of expressing reporter genes in skeletal muscle. Specifically, we show that the administration of rAAV6:hPLAP vectors to the hind limb muscles of mice is associated with dose- and time-dependent macrophage recruitment, and skeletal muscle damage. Dose-dependent expression of hPLAP also led to marked activity of established pro-inflammatory IL-6/Stat3, TNFα, IKKß and JNK signaling in lysates obtained from homogenized muscles. These effects were independent of promoter type, as expression cassettes featuring hPLAP under the control of constitutive CMV and muscle-specific CK6 promoters both drove cellular responses when matched for vector dose. Importantly, the administration of rAAV6:GFP vectors did not induce muscle damage or inflammation except at the highest doses we examined, and administration of a transgene-null vector (rAAV6:MCS) did not cause damage or inflammation at any of the doses tested, demonstrating that GFP-expressing, or transgene-null vectors may be more suitable as experimental controls. The studies highlight the importance of considering the potential effects of reporter genes when designing experiments that examine gene manipulation in vivo.


Assuntos
Genes Reporter/genética , Inflamação/patologia , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Transdução Genética , Fosfatase Alcalina/administração & dosagem , Fosfatase Alcalina/genética , Animais , Citomegalovirus/genética , Dependovirus/genética , Proteínas Ligadas por GPI/administração & dosagem , Proteínas Ligadas por GPI/genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Injeções Intramusculares , Isoenzimas/administração & dosagem , Isoenzimas/genética , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos/genética , Regiões Promotoras Genéticas/genética , Transdução de Sinais , Linfócitos T/metabolismo , Fatores de Tempo
19.
J Cell Biol ; 197(7): 997-1008, 2012 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-22711699

RESUMO

Follistatin is essential for skeletal muscle development and growth, but the intracellular signaling networks that regulate follistatin-mediated effects are not well defined. We show here that the administration of an adeno-associated viral vector expressing follistatin-288aa (rAAV6:Fst-288) markedly increased muscle mass and force-producing capacity concomitant with increased protein synthesis and mammalian target of rapamycin (mTOR) activation. These effects were attenuated by inhibition of mTOR or deletion of S6K1/2. Furthermore, we identify Smad3 as the critical intracellular link that mediates the effects of follistatin on mTOR signaling. Expression of constitutively active Smad3 not only markedly prevented skeletal muscle growth induced by follistatin but also potently suppressed follistatin-induced Akt/mTOR/S6K signaling. Importantly, the regulation of Smad3- and mTOR-dependent events by follistatin occurred independently of overexpression or knockout of myostatin, a key repressor of muscle development that can regulate Smad3 and mTOR signaling and that is itself inhibited by follistatin. These findings identify a critical role of Smad3/Akt/mTOR/S6K/S6RP signaling in follistatin-mediated muscle growth that operates independently of myostatin-driven mechanisms.


Assuntos
Folistatina/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Miostatina/metabolismo , Proteína Smad3/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Folistatina/genética , Células HEK293 , Humanos , Hipertrofia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miostatina/genética , Transdução de Sinais
20.
Methods Mol Biol ; 611: 103-14, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19960325

RESUMO

Glycoconjugates are complex macromolecules present in all tissues throughout the body. Depending on the tissue region, glycoconjugates express different carbohydrate moieties, which can be used to both distinguish cell type and examine changes in cell phenotype.Although the periodic acid-schiff (PAS) method has long been used to study the distribution of glycoconjugates, the usefulness of the technique is severely limited by its lack of specificity. A more specific technique makes use of the affinity that plant-derived lectins have for different carbohydrate moieties in glycoconjugates. Binding of lectins is therefore a particularly useful adjunct to conventional histology when it is important to characterise cell type. These well-characterised binding patterns have proved particularly valuable in helping us understand the pathogenesis of kidney disease, changes in cell surface carbohydrates on normal and neoplastic cells in tumours, and blood group biology.When labeled with a reporter molecule such as biotin or gold, lectin binding can be easily identified using light and electron microscopy. In this chapter, we describe the appropriate experimental protocols for light and electron microscopic examination of lectin binding, emphasising their utility in characterising nephron segments in renal disease.


Assuntos
Histocitoquímica/métodos , Lectinas/química , Microscopia Eletrônica/métodos , Microscopia/métodos , Animais , Glicoconjugados/química , Técnicas In Vitro , Inclusão em Parafina , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA