Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
PLoS Biol ; 20(5): e3001660, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35594316

RESUMO

In polarized epithelial cells, receptor-ligand interactions can be restricted by different spatial distributions of the 2 interacting components, giving rise to an underappreciated layer of regulatory complexity. We explored whether such regulation occurs in the Drosophila wing disc, an epithelial tissue featuring the TGF-ß family member Decapentaplegic (Dpp) as a morphogen controlling growth and patterning. Dpp protein has been observed in an extracellular gradient within the columnar cell layer of the disc, but also uniformly in the disc lumen, leading to the question of how graded signaling is achieved in the face of 2 distinctly localized ligand pools. We find the Dpp Type II receptor Punt, but not the Type I receptor Tkv, is enriched at the basolateral membrane and depleted at the junctions and apical surface. Wit, a second Type II receptor, shows a markedly different behavior, with the protein detected on all membrane regions but enriched at the apical side. Mutational studies identified a short juxtamembrane sequence required for basolateral restriction of Punt in both wing discs and mammalian Madin-Darby canine kidney (MDCK) cells. This basolateral targeting (BLT) determinant can dominantly confer basolateral localization on an otherwise apical receptor. Rescue of punt mutants with transgenes altered in the targeting motif showed that flies expressing apicalized Punt due to the lack of a functional BLT displayed developmental defects, female sterility, and significant lethality. We also show that apicalized Punt does not produce an ectopic signal, indicating that the apical pool of Dpp is not a significant signaling source even when presented with Punt. Instead, we find that basolateral presentation of Punt is required for optimal signaling. Finally, we present evidence that the BLT acts through polarized sorting machinery that differs between types of epithelia. This suggests a code whereby each epithelial cell type may differentially traffic common receptors to enable distinctive responses to spatially localized pools of extracellular ligands.


Assuntos
Proteínas de Drosophila , Drosophila , Fator de Crescimento Transformador beta , Animais , Membrana Celular/metabolismo , Cães , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Feminino , Ligantes , Células Madin Darby de Rim Canino , Proteínas Serina-Treonina Quinases , Receptores de Superfície Celular/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
2.
Cells ; 10(12)2021 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-34944010

RESUMO

Idiopathic pulmonary fibrosis (IPF) remains an intractably fatal disorder, despite the recent advent of anti-fibrotic medication. Successful treatment of IPF, like many chronic diseases, may benefit from the concurrent use of multiple agents that exhibit synergistic benefit. In this light, phosphodiesterase type 5 inhibitors (PDE5-Is), have been studied in IPF primarily for their established pulmonary vascular effects. However, recent data suggest certain PDE5-Is, particularly vardenafil, may also reduce transforming growth factor beta 1 (TGF-ß1) activation and extracellular matrix (ECM) accumulation, making them a potential target for therapy for IPF. We evaluated fibroblast TGF-ß1-driven extracellular matrix (ECM) generation and signaling as well as epithelial mesenchymal transformation (EMT) with pretreatment using the PDE5-I vardenafil. In addition, combinations of vardenafil and nintedanib were evaluated for synergistic suppression of EMC using a fibronectin enzyme-linked immunosorbent assay (ELISA). Finally, the effects of vardenafil on fibrosis were investigated in a bleomycin mouse model. Our findings demonstrate that vardenafil suppresses ECM generation alone and also exhibits significant synergistic suppression of ECM in combination with nintedanib in vitro. Interestingly, vardenafil was shown to improve fibrosis markers and increase survival in bleomycin-treated mice. Vardenafil may represent a potential treatment for IPF alone or in combination with nintedanib. However, additional studies will be required.


Assuntos
Fibrose Pulmonar Idiopática/tratamento farmacológico , Indóis/uso terapêutico , Dicloridrato de Vardenafila/uso terapêutico , Animais , Bleomicina , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Sinergismo Farmacológico , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/patologia , Indóis/farmacologia , Pulmão/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 2 de Rapamicina/metabolismo , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Análise de Sobrevida , Fator de Crescimento Transformador beta1/metabolismo , Dicloridrato de Vardenafila/farmacologia
3.
FASEB J ; 34(7): 8920-8940, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32519817

RESUMO

In the current work we show that the profibrotic actions of TGF-ß are mediated, at least in part, through a metabolic maladaptation in glutamine metabolism and how the inhibition of glutaminase 1 (GLS1) reverses pulmonary fibrosis. GLS1 was found to be highly expressed in fibrotic vs normal lung fibroblasts and the expression of profibrotic targets, cell migration, and soft agar colony formation stimulated by TGF-ß required GLS1 activity. Moreover, knockdown of SMAD2 or SMAD3 as well as inhibition of PI3K, mTORC2, and PDGFR abrogated the induction of GLS1 by TGF-ß. We further demonstrated that the NAD-dependent protein deacetylase, SIRT7, and the FOXO4 transcription factor acted as endogenous brakes for GLS1 expression, which are inhibited by TGF-ß. Lastly, administration of the GLS1 inhibitor CB-839 attenuated bleomycin-induced pulmonary fibrosis. Our study points to an exciting and unexplored connection between epigenetic and transcriptional processes that regulate glutamine metabolism and fibrotic development in a TGF-ß-dependent manner.


Assuntos
Fibroblastos/patologia , Regulação da Expressão Gênica , Glutaminase/metabolismo , Fibrose Pulmonar/patologia , Sirtuínas/metabolismo , Fator de Crescimento Transformador beta/toxicidade , Animais , Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Movimento Celular , Células Cultivadas , Feminino , Fibroblastos/metabolismo , Glutaminase/genética , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Fibrose Pulmonar/metabolismo , Transdução de Sinais , Sirtuínas/genética , Proteínas Smad/genética , Proteínas Smad/metabolismo
4.
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
5.
FASEB J ; 34(2): 2213-2226, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31907984

RESUMO

Transforming growth factor-beta (TGFß) is an enigmatic protein with various roles in healthy tissue homeostasis/development as well as the development or progression of cancer, wound healing, fibrotic disorders, and immune modulation, to name a few. As TGFß is causal to various fibroproliferative disorders featuring localized or systemic tissue/organ fibrosis as well as the activated stroma observed in various malignancies, characterizing the pathways and players mediating its action is fundamental. In the current study, we found that TGFß induces the expression of the immunoinhibitory molecule Programed death-ligand 1 (PD-L1) in human and murine fibroblasts in a Smad2/3- and YAP/TAZ-dependent manner. Furthermore, PD-L1 knockdown decreased the TGFß-dependent induction of extracellular matrix proteins, including collagen Iα1 (colIα1) and alpha-smooth muscle actin (α-SMA), and cell migration/wound healing. In addition to an endogenous role for PD-L1 in profibrotic TGFß signaling, TGFß stimulated-human lung fibroblast-derived PD-L1 into extracellular vesicles (EVs) capable of inhibiting T cell proliferation in response to T cell receptor stimulation and mediating fibroblast cell migration. These findings provide new insights and potential targets for a variety of fibrotic and malignant diseases.


Assuntos
Antígeno B7-H1/biossíntese , Vesículas Extracelulares/metabolismo , Fibroblastos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Células 3T3 , Animais , Antígeno B7-H1/genética , Vesículas Extracelulares/patologia , Fibroblastos/patologia , Fibrose , Regulação da Expressão Gênica , Humanos , Camundongos , Fator de Crescimento Transformador beta/genética
6.
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
7.
PLoS One ; 14(9): e0222083, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31483844

RESUMO

B7-1 proteins are routinely expressed on the surface of antigen presenting cells (APC) and within the innate immune system. They function to establish a biologically optimal and dynamic balance between immune activation and inhibition or self-tolerance. Interactions between B7-1 and its receptors, which include CD28, CTLA4 and PD-L1, contribute to both stimulatory as well as inhibitory or homeostatic regulation. In the current study, we investigated whether the tumor-promoting actions of transforming growth factor beta (TGF-ß) disrupted this equilibrium in pancreatic cancer to promote malignant progression and an enhanced means to evade immune detection. The data show that B7-1 is (i) upregulated following treatment of pancreatic carcinoma cells with TGF-ß; (ii) induced by TGF-ß via both Smad2/3-dependent and independent pathways; (iii) required for pancreatic tumor cell in vitro migration/invasion; and (iv) necessary for TGF-ß regulated epithelial-mesenchymal transition (EMT) through induction of Snail family members. Results from the proposed studies provide valuable insights into mechanisms whereby TGF-ß regulates both the innate immune response and intrinsic properties of pancreatic tumor growth.


Assuntos
Antígeno B7-1/imunologia , Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pancreáticas/patologia , Fator de Crescimento Transformador beta/farmacologia , Linhagem Celular Tumoral , Movimento Celular/imunologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/imunologia , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Imunomodulação/efeitos dos fármacos , Invasividade Neoplásica , Proteínas Smad/metabolismo , Neoplasias Pancreáticas
8.
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
9.
Sci Rep ; 7(1): 14298, 2017 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-29085001

RESUMO

Venous neointimal hyperplasia (VNH) at the outflow vein of hemodialysis AVF is a major factor contributing to failure. CorMatrix is an extracellular matrix that has been used in cardiovascular procedures primarily as scaffolding during surgery. In the present study, we sought to determine whether CorMatrix wrapped around the outflow vein of arteriovenous fistula (AVF) at the time of creation could reduce VNH. In mice, the carotid artery to the ipsilateral jugular vein was connected to create an AVF, and CorMatrix scaffold was wrapped around the outflow vein compared to control mice that received no scaffolding. Immunohistochemistry, Western blot, and qRT-PCR were performed on the outflow vein at 7 and 21 days after AVF creation. In outflow veins treated with CorMatrix, there was an increase in the mean lumen vessel area with a decrease in the ratio of neointima area/media + adventitia area (P < 0.05). Furthermore, there was a significant increase in apoptosis, with a reduction in cell density and proliferation in the outflow veins treated with CorMatrix compared to controls (P < 0.05). Immunohistochemical analysis revealed a significant reduction in fibroblasts, myofibroblasts, macrophages, and leukocytes with a reduction in Tnf-α gene expression (P < 0.05). In conclusion, outflow veins treated with CorMatrix have reduced VNH.


Assuntos
Túnica Adventícia/patologia , Fístula Arteriovenosa/patologia , Fístula Arteriovenosa/prevenção & controle , Hiperplasia/prevenção & controle , Diálise Renal/efeitos adversos , Alicerces Teciduais , Animais , Apoptose/fisiologia , Artérias Carótidas/cirurgia , Proliferação de Células/fisiologia , Matriz Extracelular/fisiologia , Fibroblastos/citologia , Veias Jugulares/cirurgia , Leucócitos/citologia , Macrófagos/citologia , Camundongos , Camundongos Transgênicos , Modelos Animais , Miofibroblastos/citologia , Insuficiência Renal Crônica/terapia , Fator de Necrose Tumoral alfa/análise
10.
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
11.
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
12.
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
13.
J Vasc Interv Radiol ; 27(4): 576-83, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26948326

RESUMO

PURPOSE: To determine if a second dose of a lentivirus mediated small hairpin RNA that inhibits Vegf-A gene expression (LV-shRNA-Vegf-A) can improve lumen vessel area (LVA) of the outflow vein of an arteriovenous fistula (AVF) and decrease venous neointimal hyperplasia. MATERIALS AND METHODS: Chronic kidney disease was created in C57BL/6 mice; 28 days later, an AVF was created by connecting the right carotid artery to the ipsilateral jugular vein. Immediately after AVF creation, 5 × 10(6) plaque-forming units of LV-shRNA-Vegf-A or control shRNA was administered to the adventitia of the outflow vein, and a second dose of the same treatment was administered 14 days later. Animals were sacrificed at 21 days, 28 days, and 42 days after AVF creation for reverse transcription polymerase chain reaction and histomorphometric analyses. RESULTS: By day 21, there was a 125% increase in the average LVA (day 21, P = .11), with a decrease in cell proliferation (day 21, P = .0079; day 28, P = .28; day 42, P = .5), decrease in α-smooth muscle cell actin staining (day 21, P < .0001; day 28, P < .05; day 42, P = .59), and decrease in hypoxic stress (day 21, P < .001; day 28, P = .28; day 42, P = .46) in LV versus control shRNA vessels. CONCLUSIONS: A second dose of LV-shRNA-Vegf-A administration results in a moderate improvement in LVA at day 21.


Assuntos
Derivação Arteriovenosa Cirúrgica/efeitos adversos , Vetores Genéticos , Oclusão de Enxerto Vascular/prevenção & controle , Veias Jugulares/cirurgia , Lentivirus/genética , RNA Interferente Pequeno/genética , Terapêutica com RNAi/métodos , Insuficiência Renal Crônica/terapia , Fator A de Crescimento do Endotélio Vascular/genética , Actinas/metabolismo , Túnica Adventícia/metabolismo , Animais , Apoptose , Artérias Carótidas/cirurgia , Proliferação de Células , Modelos Animais de Doenças , Oclusão de Enxerto Vascular/genética , Oclusão de Enxerto Vascular/metabolismo , Oclusão de Enxerto Vascular/patologia , Veias Jugulares/metabolismo , Veias Jugulares/patologia , Lentivirus/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Neointima , RNA Interferente Pequeno/metabolismo , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Remodelação Vascular
14.
Radiology ; 279(2): 513-22, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26583911

RESUMO

PURPOSE: To determine if adventitial transplantation of human adipose tissue-derived mesenchymal stem cells (MSCs) to the outflow vein of B6.Cg-Foxn1(nu)/J mice with arteriovenous fistula (AVF) at the time of creation would reduce monocyte chemoattractant protein-1 (Mcp-1) gene expression and venous neointimal hyperplasia. The second aim was to track transplanted zirconium 89 ((89)Zr)-labeled MSCs serially with positron emission tomography (PET) for 21 days. MATERIALS AND METHODS: All animal experiments were performed according to protocols approved by the institutional animal care and use committee. Fifty B6.Cg-Foxn1(nu)/J mice were used to accomplish the study aims. Green fluorescent protein was used to stably label 2.5 × 10(5) MSCs, which were injected into the adventitia of the outflow vein at the time of AVF creation in the MSC group. Eleven mice died after AVF placement. Animals were sacrificed on day 7 after AVF placement for real-time polymerase chain reaction (n = 6 for MSC and control groups) and histomorphometric (n = 6 for MSC and control groups) analyses and on day 21 for histomorphometric analysis only (n = 6 for MSC and control groups). In a separate group of experiments (n = 3), animals with transplanted (89)Zr-labeled MSCs were serially imaged with PET for 3 weeks. Multiple comparisons were performed with two-way analysis of variance, followed by the Student t test with post hoc Bonferroni correction. RESULTS: In vessels with transplanted MSCs compared with control vessels, there was a significant decrease in Mcp-1 gene expression (day 7: mean reduction, 62%; P = .029), with a significant increase in the mean lumen vessel area (day 7: mean increase, 176% [P = .013]; day 21: mean increase, 415% [P = .011]). Moreover, this was accompanied by a significant decrease in Ki-67 index (proliferation on day 7: mean reduction, 81% [P = .0003]; proliferation on day 21: mean reduction, 60%, [P = .016]). Prolonged retention of MSCs at the adventitia was evidenced by serial PET images of (89)Zr-labeled cells. CONCLUSION: Adventitial transplantation of MSCs decreases Mcp-1 gene expression, accompanied by a reduction in venous neointimal hyperplasia.


Assuntos
Derivação Arteriovenosa Cirúrgica/efeitos adversos , Transplante de Células-Tronco Mesenquimais , Neointima/patologia , Tecido Adiposo/citologia , Animais , Humanos , Hiperplasia/patologia , Hiperplasia/prevenção & controle , Marcação In Situ das Extremidades Cortadas , Camundongos , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Reação em Cadeia da Polimerase em Tempo Real
15.
Mol Biol Cell ; 26(21): 3879-91, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26337383

RESUMO

Transforming growth factor ß (TGFß) is a pleiotropic protein secreted from essentially all cell types and primary tissues. While TGFß's actions reflect the activity of a number of signaling networks, the primary mediator of TGFß responses are the Smad proteins. Following receptor activation, these cytoplasmic proteins form hetero-oligomeric complexes that translocate to the nucleus and affect gene transcription. Here, through biological, biochemical, and immunofluorescence approaches, sorting nexin 9 (SNX9) is identified as being required for Smad3-dependent responses. SNX9 interacts with phosphorylated (p) Smad3 independent of Smad2 or Smad4 and promotes more rapid nuclear delivery than that observed independent of ligand. Although SNX9 does not bind nucleoporins Nup153 or Nup214 or some ß importins (Imp7 or Impß), it mediates the association of pSmad3 with Imp8 and the nuclear membrane. This facilitates nuclear translocation of pSmad3 but not SNX9.


Assuntos
Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Nexinas de Classificação/genética , Nexinas de Classificação/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Técnicas de Cultura de Células , Núcleo Celular/metabolismo , Humanos , Carioferinas/metabolismo , Ligantes , Camundongos , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Fosforilação , Ligação Proteica , Transporte Proteico , Transdução de Sinais , Proteínas Smad/metabolismo , Proteína Smad4/metabolismo , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 35(6): 1401-12, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25882068

RESUMO

OBJECTIVE: Neuropilin-1 (NRP-1) is a multidomain membrane receptor involved in angiogenesis and development of neuronal circuits, however, the role of NRP-1 in cardiovascular pathophysiology remains elusive. APPROACH AND RESULTS: In this study, we first observed that deletion of NRP-1 induced peroxisome proliferator-activated receptor γ coactivator 1α in cardiomyocytes and vascular smooth muscle cells, which was accompanied by dysregulated cardiac mitochondrial accumulation and induction of cardiac hypertrophy- and stress-related markers. To investigate the role of NRP-1 in vivo, we generated mice lacking Nrp-1 in cardiomyocytes and vascular smooth muscle cells (SM22-α-Nrp-1 KO), which exhibited decreased survival rates, developed cardiomyopathy, and aggravated ischemia-induced heart failure. Mechanistically, we found that NRP-1 specifically controls peroxisome proliferator-activated receptor γ coactivator 1 α and peroxisome proliferator-activated receptor γ in cardiomyocytes through crosstalk with Notch1 and Smad2 signaling pathways, respectively. Moreover, SM22-α-Nrp-1 KO mice exhibited impaired physical activities and altered metabolite levels in serum, liver, and adipose tissues, as demonstrated by global metabolic profiling analysis. CONCLUSIONS: Our findings provide new insights into the cardioprotective role of NRP-1 and its influence on global metabolism.


Assuntos
Cardiomiopatias/metabolismo , Insuficiência Cardíaca/metabolismo , Isquemia Miocárdica/metabolismo , Neuropilina-1/metabolismo , Animais , Homeostase , Camundongos Knockout , Proteínas dos Microfilamentos , Mitocôndrias Cardíacas/metabolismo , Proteínas Musculares , Músculo Liso Vascular/metabolismo , Miócitos Cardíacos/metabolismo , PPAR gama/metabolismo , Receptor Cross-Talk , Receptor Notch1/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Fatores de Transcrição/metabolismo
17.
Dev Cell ; 32(5): 640-51, 2015 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-25758862

RESUMO

Cell-cell contacts inhibit cell growth and proliferation in part by activating the Hippo pathway that drives the phosphorylation and nuclear exclusion of the transcriptional coactivators YAP and TAZ. Cell density and Hippo signaling have also been reported to block transforming growth factor ß (TGF-ß) responses, based on the ability of phospho-YAP/TAZ to sequester TGF-ß-activated SMAD complexes in the cytoplasm. Herein, we provide evidence that epithelial cell polarization interferes with TGF-ß signaling well upstream and independent of cytoplasmic YAP/TAZ. Rather, polarized basolateral presentation of TGF-ß receptors I and II deprives apically delivered TGF-ß of access to its receptors. Basolateral ligand delivery nonetheless remains entirely effective to induce TGF-ß responses. These data demonstrate that cell-type-specific inhibition of TGF-ß signaling by cell density is restricted to polarized epithelial cells and reflects the polarized distribution of TGF-ß receptors, which thus affects SMAD activation irrespective of Hippo pathway activation.


Assuntos
Citoplasma/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Aciltransferases , Western Blotting , Contagem de Células , Proteínas de Ciclo Celular , Proliferação de Células , Células Cultivadas , Imunofluorescência , Via de Sinalização Hippo , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Proteínas Nucleares/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta/genética
18.
J Vasc Res ; 52(5): 334-46, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26985676

RESUMO

PURPOSE: It is hypothesized that venous stenosis formation associated with hemodialysis vascular-access failure is caused by hypoxia-mediated fibroblast-to-myofibroblast differentiation accompanied by proliferation and migration, and that diabetic patients have worse clinical outcomes. The aim of this study was to determine the functional and gene expression outcomes of matrix metalloproteinase-2 (Mmp-2) silencing in fibroblasts cultured under hyperglycemia and euglycemia with hypoxic and normoxic stimuli. MATERIALS AND METHODS: AKR-2B fibroblasts were stably transduced using lentivirus-mediated shRNA-Mmp-2 or scrambled controls and subjected to hypoxia or normoxia under hyperglycemic or euglycemic conditions for 24 and 72 h. Gene expression of vascular endothelial growth factor-A (Vegf-A), Vegfr-1, Mmp-2, Mmp-9 and tissue inhibitors of matrix metalloproteinases (Timps) were determined by RT-PCR. Collagen I and IV secretion and cellular proliferation and migration were determined. RESULTS: Under hyperglycemic conditions, there is a significant reduction in the average gene expression of Vegf-A and Mmp-9, with an increase in Timp-1 at 24 h of hypoxia (p < 0.05) in Mmp-2-silenced fibroblasts when compared to controls. In addition, there is a decrease in collagen I and IV secretion and cellular migration. The euglycemic cells were able to reverse these findings. CONCLUSION: These findings demonstrate the rationale for using anti-Mmp-2 therapy in dialysis patients with hemodialysis vascular access in helping to reduce stenosis formation.


Assuntos
Derivação Arteriovenosa Cirúrgica/efeitos adversos , Angiopatias Diabéticas/enzimologia , Fibroblastos/enzimologia , Glucose/metabolismo , Oclusão de Enxerto Vascular/enzimologia , Metaloproteinase 2 da Matriz/metabolismo , Neovascularização Patológica , Diálise Renal , Animais , Hipóxia Celular , Linhagem Celular , Movimento Celular , Proliferação de Células , Colágeno Tipo I/metabolismo , Colágeno Tipo IV/metabolismo , Meios de Cultivo Condicionados/metabolismo , Angiopatias Diabéticas/genética , Angiopatias Diabéticas/patologia , Fibroblastos/patologia , Regulação da Expressão Gênica , Oclusão de Enxerto Vascular/genética , Oclusão de Enxerto Vascular/patologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Miofibroblastos/enzimologia , Miofibroblastos/patologia , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Inibidores Teciduais de Metaloproteinases/genética , Inibidores Teciduais de Metaloproteinases/metabolismo , Transfecção , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
19.
Kidney Int ; 85(2): 289-306, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23924957

RESUMO

Venous neointimal hyperplasia (VNH) causes hemodialysis vascular access failure. Here we tested whether VNH formation occurs in part due to local vessel hypoxia caused by surgical trauma to the vasa vasorum of the outflow vein at the time of arteriovenous fistula placement. Selective targeting of the adventitia of the outflow vein at the time of fistula creation was performed using a lentivirus-delivered small-hairpin RNA that inhibits VEGF-A expression. This resulted in significant increase in mean lumen vessel area, decreased media/adventitia area, and decreased constrictive remodeling with a significant increase in apoptosis (increase in caspase 3 activity and TUNEL staining) accompanied with decreased cellular proliferation and hypoxia-inducible factor-1α at the outflow vein. There was significant decrease in cells staining positive for α-smooth muscle actin (a myofibroblast marker) and VEGFR-1 expression with a decrease in MMP-2 and MMP-9. These results were confirmed in animals that were treated with humanized monoclonal antibody to VEGF-A with similar results. Since hypoxia can cause fibroblast to differentiate into myofibroblasts, we silenced VEGF-A gene expression in fibroblasts and subjected them to hypoxia. This decreased myofibroblast production, cellular proliferation, cell invasion, MMP-2 activity, and increased caspase 3. Thus, VEGF-A reduction at the time of arteriovenous fistula placement results in increased positive vascular remodeling.


Assuntos
Túnica Adventícia/cirurgia , Derivação Arteriovenosa Cirúrgica/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos , Oclusão de Enxerto Vascular/prevenção & controle , Veias Jugulares/cirurgia , Lentivirus/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução Genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Túnica Adventícia/metabolismo , Túnica Adventícia/patologia , Animais , Apoptose , Artérias Carótidas/cirurgia , Caspase 3/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Constrição Patológica , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibroblastos/patologia , Oclusão de Enxerto Vascular/genética , Oclusão de Enxerto Vascular/metabolismo , Oclusão de Enxerto Vascular/patologia , Hiperplasia , Veias Jugulares/metabolismo , Veias Jugulares/patologia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neointima , Nefrectomia , RNA Interferente Pequeno/genética , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/genética
20.
FASEB J ; 27(11): 4444-54, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23913859

RESUMO

Transforming growth factor ß (TGFß) has significant profibrotic activity both in vitro and in vivo. This reflects its capacity to stimulate fibrogenic mediators and induce the expression of other profibrotic cytokines such as platelet-derived growth factor (PDGF) and epidermal growth factor (EGF/ErbB) ligands. Here we address both the mechanisms by which TGFß induced ErbB ligands and the physiological significance of inhibiting multiple TGFß-regulated processes. The data document that ErbB ligand induction requires PDGF receptor (PDGFR) mediation and engages a positive autocrine/paracrine feedback loop via ErbB receptors. Whereas PDGFRs are essential for TGFß-stimulated ErbB ligand up-regulation, TGFß-specific signals are also required for ErbB receptor activation. Subsequent profibrotic responses are shown to involve the cooperative action of PDGF and ErbB signaling. Moreover, using a murine treatment model of bleomycin-induced pulmonary fibrosis we found that inhibition of TGFß/PDGF and ErbB pathways with imatinib plus lapatinib, respectively, not only prevented myofibroblast gene expression to a greater extent than either drug alone, but also essentially stabilized gas exchange (oxygen saturation) as an overall measure of lung function. These observations provide important mechanistic insights into profibrotic TGFß signaling and indicate that targeting multiple cytokines represents a possible strategy to ameliorate organ fibrosis dependent on TGFß.


Assuntos
Receptores ErbB/metabolismo , Fibrose Pulmonar/metabolismo , Receptor ErbB-2/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Animais , Benzamidas/administração & dosagem , Benzamidas/uso terapêutico , Bleomicina/toxicidade , Linhagem Celular , Interações Medicamentosas , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/metabolismo , Retroalimentação Fisiológica , Mesilato de Imatinib , Lapatinib , Pulmão/fisiopatologia , Camundongos , Miofibroblastos/metabolismo , Comunicação Parácrina , Piperazinas/administração & dosagem , Piperazinas/uso terapêutico , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico , Troca Gasosa Pulmonar , Pirimidinas/administração & dosagem , Pirimidinas/uso terapêutico , Quinazolinas/administração & dosagem , Quinazolinas/uso terapêutico , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...