Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 34(4): 5363-5388, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32067272

RESUMO

Pathogenic fibrotic diseases, including idiopathic pulmonary fibrosis (IPF), have some of the worst prognoses and affect millions of people worldwide. With unclear etiology and minimally effective therapies, two-thirds of IPF patients die within 2-5 years from this progressive interstitial lung disease. Transforming Growth Factor Beta (TGFß) and insulin-like growth factor-1 (IGF-1) are known to promote fibrosis; however, myofibroblast specific upregulation of IGF-1 in the initiation and progression of TGFß-induced fibrogenesis and IPF have remained unexplored. To address this, the current study (1) documents the upregulation of IGF-1 via TGFß in myofibroblasts and fibrotic lung tissue, as well as its correlation with decreased pulmonary function in advanced IPF; (2) identifies IGF-1's C1 promoter as mediating the increase in IGF-1 transcription by TGFß in pulmonary fibroblasts; (3) determines that SMAD2 and mTOR signaling are required for TGFß-dependent Igf-1 expression in myofibroblasts; (4) demonstrates IGF-1R activation is essential to support TGFß-driven profibrotic myofibroblast functions and excessive wound healing; and (5) establishes the effectiveness of slowing the progression of murine lung fibrosis with the IGF-1R inhibitor OSI-906. These findings expand our knowledge of IGF-1's role as a novel fibrotic-switch, bringing us one step closer to understanding the complex biological mechanisms responsible for fibrotic diseases and developing effective therapies.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Fibroblastos/patologia , Fibrose Pulmonar Idiopática/patologia , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Diferenciação Celular , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Fator de Crescimento Insulin-Like I/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL
2.
Sci Signal ; 12(612)2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31848318

RESUMO

Metabolic dysregulation in fibroblasts is implicated in the profibrotic actions of transforming growth factor-ß (TGF-ß). Here, we present evidence that hexokinase 2 (HK2) is important for mediating the fibroproliferative activity of TGF-ß both in vitro and in vivo. Both Smad-dependent and Smad-independent TGF-ß signaling induced HK2 accumulation in murine and human lung fibroblasts through induction of the transcription factor c-Myc. Knockdown of HK2 or pharmacological inhibition of HK2 activity with Lonidamine decreased TGF-ß-stimulated fibrogenic processes, including profibrotic gene expression, cell migration, colony formation, and activation of the transcription factors YAP and TAZ, with no apparent effect on cellular viability. Fibroblasts from patients with idiopathic pulmonary fibrosis (IPF) exhibited an increased abundance of HK2. In a mouse model of bleomycin-induced lung fibrosis, Lonidamine reduced the expression of genes encoding profibrotic markers (collagenΙα1, EDA-fibronectin, α smooth muscle actin, and connective tissue growth factor) and stabilized or improved lung function as assessed by measurement of peripheral blood oxygenation. These findings provide evidence of how metabolic dysregulation through HK2 can be integrated within the context of profibrotic TGF-ß signaling.


Assuntos
Glicólise , Hexoquinase/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Células 3T3 , Animais , Fibrose , Hexoquinase/genética , Camundongos , Fator de Crescimento Transformador beta/genética
3.
FASEB J ; 32(7): 3803-3815, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29475397

RESUMO

Evidence is provided that the fibroproliferative actions of TGF-ß are dependent on a metabolic adaptation that sustains pathologic growth. Specifically, profibrotic TGF-ß signaling is shown to require fatty acid synthase (FASN), an essential anabolic enzyme responsible for the de novo synthesis of fatty acids. With the use of pharmacologic and genetic approaches, we show that TGF-ß-stimulated FASN expression is independent of Smad2/3 and is mediated via mammalian target of rapamycin complex 1. In the absence of FASN activity or protein, TGF-ß-driven fibrogenic processes are reduced with no apparent toxicity. Furthermore, as increased FASN expression was also observed to correlate with the degree of lung fibrosis in bleomycin-treated mice, inhibition of FASN was examined in a murine-treatment model of pulmonary fibrosis. Remarkably, inhibition of FASN not only decreased expression of profibrotic targets, but lung function was also stabilized/improved, as assessed by peripheral blood oxygenation.-Jung, M.-Y., Kang, J.-H., Hernandez, D. M., Yin, X., Andrianifahanana, M., Wang, Y., Gonzalez-Guerrico, A., Limper, A. H., Lupu, R., Leof, E. B. Fatty acid synthase is required for profibrotic TGF-ß signaling.


Assuntos
Ácido Graxo Sintase Tipo I/metabolismo , Fibrose Pulmonar/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Bleomicina/toxicidade , Linhagem Celular , Ácido Graxo Sintase Tipo I/genética , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/etiologia , Transdução de Sinais , Proteínas Smad/metabolismo
4.
Mol Biol Cell ; 28(20): 2701-2711, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28768825

RESUMO

Delivery of biomolecules to the correct subcellular locales is critical for proper physiological function. To that end, we have previously determined that type I and II transforming growth factor beta (TGF-ß) receptors (TßRI and TßRII, respectively) localize to the basolateral domain in polarized epithelia. While TßRII targeting was shown to be regulated by sequences between amino acids 529 and 538, the analogous region(s) within TßRI is unknown. To address that question, sequential cytoplasmic TßRI truncations and point mutations identified a targeting motif between residues 158 and 163 (VxxEED) required for basolateral TßRI expression. Further studies documented that receptor internalization, down-regulation, direct recycling, or Smad signaling were unaffected by motif mutations that caused TßRI mislocalization. However, inclusion of amino acids 148-217 containing the targeting motif was able to direct basolateral expression of the apically sorted nerve growth factor receptor (NGFR, p75; extracellular and transmembrane regions) in a dominant manner. Finally, coexpression of apically targeted type I and type II TGF-ß receptors mediated Smad3 signaling from the apical membrane of polarized epithelial cells. These findings demonstrate that the absence of apical TGF-ß signaling in normal epithelia is primarily a reflection of domain-specific receptor expression and not an inability to couple with the signaling machinery.


Assuntos
Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Motivos de Aminoácidos , Animais , Membrana Celular/metabolismo , Polaridade Celular/fisiologia , Citoplasma/metabolismo , Cães , Expressão Gênica , Humanos , Células Madin Darby de Rim Canino , Ligação Proteica , Transporte Proteico , Receptor do Fator de Crescimento Transformador beta Tipo II , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
5.
J Clin Invest ; 127(7): 2541-2554, 2017 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-28530637

RESUMO

TGF-ß is considered a master switch in the pathogenesis of organ fibrosis. The primary mediators of this activity are the SMAD proteins, particularly SMAD3. In the current study, we have developed a cell-penetrating peptide (CPP) conjugate of the HIV TAT protein that is fused to an aminoterminal sequence of sorting nexin 9 (SNX9), which was previously shown to bind phosphorylated SMAD3 (pSMAD3). We determined that specifically preventing the nuclear import of pSMAD3 using the TAT-SNX9 peptide inhibited profibrotic TGF-ß activity in murine cells and human lung fibroblasts as well as in vivo with no demonstrable toxicity. TGF-ß signaling mediated by pSMAD2, bone morphogenetic protein 4 (BMP4), EGF, or PDGF was unaffected by the TAT-SNX9 peptide. Furthermore, while the TAT-SNX9 peptide prevented TGF-ß's profibrotic activity in vitro as well as in 2 murine treatment models of pulmonary fibrosis, a 3-amino acid point mutant that was unable to bind pSMAD3 proved ineffective. These findings indicate that specifically targeting pSMAD3 can ameliorate both the direct and indirect fibroproliferative actions of TGF-ß.


Assuntos
Peptídeos Penetradores de Células/farmacologia , Fibrose Pulmonar/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/metabolismo , Animais , Linhagem Celular , Peptídeos Penetradores de Células/genética , Modelos Animais de Doenças , Feminino , Camundongos , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Transdução de Sinais/genética , Proteína Smad3/genética , Fator de Crescimento Transformador beta/genética
6.
FASEB J ; 30(11): 3733-3744, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27480571

RESUMO

TGF-ß plays a central role in the pathogenesis of fibroproliferative disorders. Defining the exact underlying molecular basis is therefore critical for the development of viable therapeutic strategies. Here, we show that expression of the facilitative glucose transporter 1 (GLUT1) is induced by TGF-ß in fibroblast lines and primary cells and is required for the profibrotic effects of TGF-ß. In addition, enhanced GLUT1 expression is observed in fibrotic areas of lungs of both patients with idiopathic pulmonary fibrosis and mice that are subjected to a fibrosis-inducing bleomycin treatment. By using pharmacologic and genetic approaches, we demonstrate that up-regulation of GLUT1 occurs via the canonical Smad2/3 pathway and requires autocrine activation of the receptor tyrosine kinases, platelet-derived and epidermal growth factor receptors. Engagement of the common downstream effector PI3K subsequently triggers activation of the MEK and mammalian target of rapamycin complex 2, which cooperate in regulating GLUT1 expression. Of note, inhibition of GLUT1 activity and/or expression is shown to impair TGF-ß-driven fibrogenic processes, including cell proliferation and production of profibrotic mediators. These findings provide new perspectives on the interrelation of metabolism and profibrotic TGF-ß signaling and present opportunities for potential therapeutic intervention.-Andrianifahanana, M., Hernandez, D. M., Yin, X., Kang, J.-H., Jung, M.-Y., Wang, Y., Yi, E. S., Roden, A. C., Limper, A. H., Leof, E. B. Profibrotic up-regulation of glucose transporter 1 by TGF-ß involves activation of MEK and mammalian target of rapamycin complex 2 pathways.


Assuntos
Proliferação de Células/fisiologia , Transportador de Glucose Tipo 1/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Sirolimo/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fibroblastos/metabolismo , Fibrose/metabolismo , Pulmão/metabolismo , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...