Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Cell Mol Med ; 24(6): 3481-3491, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32052562

RESUMO

Patients on peritoneal dialysis are at risk of developing peritoneal fibrosis and angiogenesis, which can lead to dysfunction of the peritoneal membrane. Recent evidence has identified cross-talk between transforming growth factor beta (TGFB) and the WNT/ß-catenin pathway to induce fibrosis and angiogenesis. Limited evidence exists describing the role of non-canonical WNT signalling in peritoneal membrane injury. Non-canonical WNT5A is suggested to have different effects depending on the receptor environment. WNT5A has been implicated in antagonizing canonical WNT/ß-catenin signalling in the presence of receptor tyrosine kinase-like orphan receptor (Ror2). We co-expressed TGFB and WNT5A using adenovirus and examined its role in the development of peritoneal fibrosis and angiogenesis. Treatment of mouse peritoneum with AdWNT5A decreased the submesothelial thickening and angiogenesis induced by AdTGFB. WNT5A appeared to block WNT/ß-catenin signalling by inhibiting phosphorylation of glycogen synthase kinase 3 beta (GSK3B) and reducing levels of total ß-catenin and target proteins. To examine the function of Ror2, we silenced Ror2 in a human mesothelial cell line. We treated cells with AdWNT5A and observed a significant increase in fibronectin compared with AdWNT5A alone. We also analysed fibronectin and vascular endothelial growth factor (VEGF) in a TGFB model of mesothelial cell injury. Both fibronectin and VEGF were significantly increased in response to Ror2 silencing when cells were exposed to TGFB. Our results suggest that WNT5A inhibits peritoneal injury and this is associated with a decrease in WNT/ß-catenin signalling. In human mesothelial cells, Ror2 is involved in regulating levels of fibronectin and VEGF.


Assuntos
Peritônio/metabolismo , Peritônio/patologia , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Proteína Wnt-5a/metabolismo , Animais , Transição Epitelial-Mesenquimal , Epitélio/metabolismo , Fibronectinas/metabolismo , Fibrose , Humanos , Membranas , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Fator de Crescimento Transformador beta/metabolismo , Via de Sinalização Wnt
2.
Am J Physiol Renal Physiol ; 314(6): F1036-F1045, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29363326

RESUMO

The wingless-type mouse mammary tumor virus integration site family (WNT) signaling pathway is involved in wound healing and fibrosis. We evaluated the WNT signaling pathway in peritoneal membrane injury. We assessed WNT1 protein expression in the peritoneal effluents of 54 stable peritoneal dialysis (PD) patients and WNT-related gene expression in ex vivo mesothelial cell cultures from 21 PD patients. In a transforming growth factor-ß (TGF-ß)-mediated animal model of peritoneal fibrosis, we evaluated regulation of the WNT pathway and the effect of WNT inhibition on peritoneal fibrosis and angiogenesis. WNT1 and WNT2 gene expression were positively correlated with peritoneal membrane solute transport in PD patients. In the mouse peritoneum, TGF-ß-induced peritoneal fibrosis was associated with increased expression of WNT2 and WNT4. Peritoneal ß-catenin protein was significantly upregulated after infection with adenovirus expressing TGF-ß (AdTGF-ß) along with elements of the WNT signaling pathway. Treatment with a ß-catenin inhibitor (ICG-001) in mice with AdTGF-ß-induced peritoneal fibrosis resulted in attenuation of peritoneal angiogenesis and reduced vascular endothelial growth factor. Similar results were also observed with the WNT antagonist Dickkopf-related protein (DKK)-1. In addition to this, DKK-1 blocked epithelial-mesenchymal transition and increased levels of the cell adhesion protein E-cadherin. We provide evidence that WNT signaling is active in the setting of experimental peritoneal fibrosis and WNT1 correlates with patient peritoneal membrane solute transport in PD patients. Intervention in this pathway is a possible therapy for peritoneal membrane injury.


Assuntos
Células Epiteliais/metabolismo , Neovascularização Patológica , Fibrose Peritoneal/metabolismo , Peritônio/irrigação sanguínea , Peritônio/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Idoso , Animais , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Diálise Peritoneal/efeitos adversos , Fibrose Peritoneal/etiologia , Fibrose Peritoneal/genética , Fibrose Peritoneal/patologia , Peritônio/patologia , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Proteínas Wnt/genética , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , Proteína Wnt4/genética , Proteína Wnt4/metabolismo , beta Catenina/metabolismo
3.
Nephrol Dial Transplant ; 32(1): 50-61, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27190383

RESUMO

Background: For patients using peritoneal dialysis (PD), the peritoneal membrane can develop fibrosis and angiogenesis, leading to ultrafiltration failure, chronic hypervolemia and increased risk of technique failure and mortality. Matrix metalloproteinases (MMPs), and specifically the gelatinases (MMP2 and MMP9), may be involved in peritoneal membrane injury. Methods: From stable PD patients, mesothelial cells were assayed for MMP gene expression. MMP9 was overexpressed in mouse peritoneum by adenovirus, and MMP9 -/- mice were subjected to transforming growth factor ß (TGF-ß)-induced peritoneal fibrosis. Results: MMP9 mRNA expression correlated with peritoneal membrane solute transport properties. Overexpression of MMP9 in the mouse peritoneum induced submesothelial thickening and angiogenesis. MMP9 induced mesothelial cell transition to a myofibroblast phenotype measured by increased alpha smooth muscle actin and decreased E-cadherin expression. Angiogenesis was markedly reduced in MMP9 -/- mice treated with an adenovirus expressing active TGF-ß compared with wild-type mice. TGF-ß-mediated E-cadherin cleavage was MMP9 dependent, and E-cadherin cleavage led to ß-catenin-mediated signaling. A ß-catenin inhibitor blocked the angiogenic response induced by AdMMP9. Conclusions: Our data suggest that MMP9 is involved in peritoneal membrane injury possibly through cleavage of E-cadherin and induction of ß-catenin signaling. MMP9 is a potential biomarker for peritoneal membrane injury and is a therapeutic target to protect the peritoneal membrane in PD patients.


Assuntos
Caderinas/metabolismo , Soluções para Hemodiálise/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Patológica/etiologia , Diálise Peritoneal/efeitos adversos , Fibrose Peritoneal/etiologia , beta Catenina/metabolismo , Animais , Transporte Biológico , Caderinas/genética , Humanos , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Fibrose Peritoneal/metabolismo , Fibrose Peritoneal/patologia , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , beta Catenina/genética
4.
Perit Dial Int ; 35(5): 517-29, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25395500

RESUMO

BACKGROUND: Outcomes for peritoneal dialysis (PD) patients are affected by the characteristics of the peritoneal membrane, which may be determined by genetic variants. We carried out a systematic review of the literature to identify studies which assessed the association between genetic polymorphisms, peritoneal membrane solute transport, and clinical outcomes for PD patients. METHODS: The National Library of Medicine was searched using a variety of strategies. Studies which met our inclusion criteria were reviewed and data abstracted. Our outcomes of interest included: high transport status peritoneal membrane, risk for peritonitis, encapsulating peritoneal sclerosis (EPS), patient and technique survival. We combined data from studies which evaluated the same genetic polymorphism and the same outcome. RESULTS: We evaluated 18 relevant studies. All studies used a candidate gene approach. Gene polymorphisms in the interleukin (IL)-6 gene were associated with peritoneal membrane solute transport in several studies in different ethnic populations. Associations with solute transport and polymorphisms in endothelial nitric oxide synthase and receptor for advanced glycation end product genes were also identified. There was evidence of a genetic predisposition for peritonitis found in 2 studies, and for EPS in 1 study. Survival was found to be associated with a polymorphism in vascular endothelial growth factor and technique failure was associated with a polymorphism in the IL-1 receptor antagonist. CONCLUSIONS: There is evidence that characteristics of the peritoneal membrane and clinical outcomes for PD patients have genetic determinants. The most consistent association was between IL-6 gene polymorphisms and peritoneal membrane solute transport.


Assuntos
Proteínas de Transporte/metabolismo , Interleucina-6/genética , Peritônio/metabolismo , Polimorfismo Genético , Transporte Biológico/fisiologia , Soluções para Diálise , Humanos , Diálise Peritoneal
5.
Am J Pathol ; 184(11): 2976-84, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25194662

RESUMO

The peritoneal membrane becomes damaged in patients on peritoneal dialysis (PD). Gremlin 1 (GREM1) inhibits bone morphogenic proteins (BMPs) and plays a role in kidney development and fibrosis. We evaluated the role of gremlin in peritoneal fibrosis and angiogenesis. In a cohort of 32 stable PD patients, GREM1 concentration in the peritoneal effluent correlated with measures of peritoneal membrane damage. AdGrem1, an adenovirus to overexpress gremlin in the mouse peritoneum, induced submesothelial thickening, fibrosis, and angiogenesis in C57BL/6 mice, which was associated with decreased expression of BMP4 and BMP7. There was evidence of mesothelial cell transition to a mesenchymal phenotype with increased α smooth muscle actin expression and suppression of E-cadherin. Some of the GREM1 effects may be reversed with recombinant BMP7 or a pan-specific transforming growth factor ß (TGF-ß) antibody. Neovascularization was not inhibited with a TGF-ß antibody, suggesting a TGF-ß-independent angiogenic mechanism. Swiss/Jackson Laboratory (SJL) mice, which are resistant to TGF-ß-induced peritoneal fibrosis, responded in a similar fashion to AdGrem1 as did C57BL/6 mice with fibrosis, angiogenesis, and mesothelial-to-mesenchymal transition. GREM1 was associated with up-regulated TGF-ß expression in both SJL and C57BL/6 mice, but SJL mice demonstrated a defective TGF-ß-induced GREM1 expression. In summary, GREM1 induces fibrosis and angiogenesis in mouse peritoneum and is associated with increased solute transport in these PD patients.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neovascularização Patológica/metabolismo , Diálise Peritoneal/efeitos adversos , Fibrose Peritoneal/metabolismo , Peritônio/metabolismo , Idoso , Animais , Transporte Biológico , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Proteína Morfogenética Óssea 7/genética , Proteína Morfogenética Óssea 7/metabolismo , Modelos Animais de Doenças , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Pessoa de Meia-Idade , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Fibrose Peritoneal/etiologia , Fibrose Peritoneal/genética , Peritônio/patologia , Fator de Crescimento Transformador beta1/metabolismo
6.
Nephrol Dial Transplant ; 28(8): 2015-27, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22785109

RESUMO

BACKGROUND: Encapsulating peritoneal sclerosis (EPS) is a rare but devastating complication of peritoneal dialysis. The etiology is unclear, but genetic predisposition may be a contributing factor. We used adenovirus-mediated gene transfer of transforming growth factor (TGF) ß1 to the peritoneum in four genetically distinct laboratory mouse strains to assess differences in fibrogenic response. METHODS: Mice from four genetic backgrounds (C57BL/6J, DBA/2J, C3H/HeJ and SJL/J) received an intraperitoneal injection of an adenovirus expressing TGFß1 (AdTGFß1) or control adenovirus (AdDL) and were assessed 4 and 10 days after infection. Submesothelial thickening, angiogenesis and gene expression were quantified from peritoneal tissue. Protein was extracted from omental tissue and assessed for collagen, E-cadherin and TGFß signaling pathway proteins. RESULTS: There was a graded response among the mouse strains to the peritoneal overexpression of TGFß1. TGFß1 induced a significant fibrogenic response in the C57BL/6J mice, whereas the SJL/J mice were resistant. The DBA/2J and the C3H/HeJ mice had intermediate responses. A similar graded response was seen in collagen protein levels in the omental tissue and in fibrosis-associated gene expression. TGFß type 1 receptor and SMAD signaling pathways appeared to be intact in all the mouse strains. CONCLUSIONS: There were significant differences in mouse strain susceptibility to peritoneal fibrosis, suggesting that genetic factors may play a role in the development of peritoneal fibrosis and possibly EPS. As early TGFß1 signaling mechanisms appear to be intact, we hypothesize that fibrosis resistance in the SJL/J mice lies further down the wound-healing cascade or in an alternate, non-SMAD pathway.


Assuntos
Nefropatias/complicações , Diálise Peritoneal/efeitos adversos , Fibrose Peritoneal/etiologia , Fator de Crescimento Transformador beta/efeitos adversos , Animais , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Fibrose Peritoneal/diagnóstico , Fibrose Peritoneal/metabolismo , Proteínas Smad/metabolismo , Especificidade da Espécie
7.
Nephrol Dial Transplant ; 27(11): 3978-81, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22798507

RESUMO

BACKGROUND: The peritoneal membrane is a vital structure for peritoneal dialysis (PD) patients. It has been increasingly recognized that the transition of the peritoneal lining mesothelial cells into a more fibroblastic phenotype is a key step in peritoneal membrane injury. METHODS: Relevant literature was reviewed and summarized. RESULTS: Epithelial-to-mesenchymal transition (EMT) is a basic cellular process that occurs in a variety of physiologic and pathologic processes. The hallmark of this process is a loss of epithelial markers, and E-cadherin is a prototypical epithelial transmembrane protein. E-cadherin expression is suppressed at many levels and the gene is regulated by a family of transcription factors. Twist is one of the lesser studied E-cadherin regulatory factors, which belongs to a larger family of basic helix-loop-helix DNA-binding proteins. In this issue of Nephrology Dialysis Transplantation, Cuixiang Li reports on in vitro experiments where the expression of Twist led to a decreased expression of E-cadherin and the evidence of EMT. In an in vivo model of dialysate exposure, Li demonstrates that Twist expression is increased in the injured peritoneal tissues. CONCLUSIONS: These important observations are the first to link Twist to mesothelial cell EMT and peritoneal membrane injury. Like most novel observations, this paper leaves many questions unanswered. Twist is only one of several transcription factors involved in EMT and how these factors interact will require further investigations. Furthermore, the question of whether Twist interacts at multiple levels in the EMT process, or simply gives an initial push to the process, is left unanswered. Finally, to bring these early significant findings to the bedside as potential therapies for PD patients will require further innovation.


Assuntos
Glicemia/metabolismo , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal , Epitélio/metabolismo , Peritônio/citologia , Proteína 1 Relacionada a Twist/biossíntese , Animais , Humanos
8.
J Artif Organs ; 15(3): 272-82, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22622710

RESUMO

Marked thickening of the peritoneum and vasculopathy in the submesothelial compact zone have been reported in long-term peritoneal dialysis patients. Bone marrow (BM)-derived cell lines are considered to be useful tools for therapy of various diseases. To clarify the role of BM-derived cells in the peritoneal fibrosis (PF) model, we analyzed several lineages of cells in the peritoneum. BM cells from green fluorescent protein (GFP) transgenic mice were transplanted into naïve C57Bl/6 mice. Chlorhexidine gluconate (CG) was injected intraperitoneally to induce PF. Immunohistochemical analysis was performed with parietal peritoneum using anti-Sca-1 or -c-Kit and -GFP antibodies. Isolated BM cells were also transplanted into the CG-stimulated peritoneum. BM-derived cells from GFP transgenic mice appeared in the submesothelium from days 14 to 42. Both GFP- and stem cell marker-positive cells were observed in the submesothelium and on the surface. Isolated c-Kit-positive cells, transplanted into the peritoneal cavity, differentiated into mesothelial cells. In this study, we investigated whether or not BM-derived cells play a role in the repair of PF and immature cells have the potential of inducing repair of the peritoneum. The findings of this study suggest a new concept for therapy of PF.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular/fisiologia , Fibrose Peritoneal/patologia , Peritônio/patologia , Animais , Células da Medula Óssea/metabolismo , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fibrose Peritoneal/metabolismo , Peritônio/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo
9.
J Cell Mol Med ; 16(8): 1934-45, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22117756

RESUMO

Patients with end-stage kidney disease on peritoneal dialysis often develop progressive scarring of the peritoneal tissues. This manifests as submesothelial thickening and is associated with increased vascularization that leads to ultrafiltration dysfunction. Hypoxia induces a characteristic series of responses including angiogenesis and fibrosis. We investigated the role of hypoxia in peritoneal membrane damage. An adenovirus expressing transforming growth factor (TGF) ß was used to induce peritoneal fibrosis. We evaluated the effect of the mTOR inhibitor rapamycin, which has been previously shown to block hypoxia-inducible factor (HIF) 1α. We also assessed the effect of HIF1α independently using an adenovirus expressing active HIF1α. To identify the TGFß1-independent effects of HIF1α, we expressed HIF1α in the peritoneum of mice lacking the TGFß signalling molecule Smad3. We demonstrate that TGFß-induced fibroproliferative tissue is hypoxic. Rapamycin did not affect the early angiogenic response, but inhibited angiogenesis and submesothelial thickening 21 days after induction of fibrosis. In primary mesothelial cell culture, rapamycin had no effect on TGFß-induced vascular endothelial growth factor (VEGF) but did suppress hypoxia-induced VEGF. HIF1α induced submesothelial thickening and angiogenesis in peritoneal tissue. The fibrogenic effects of HIF1α were Smad3 dependent. In summary, submesothelial hypoxia may be an important secondary factor, which augments TGFß-induced peritoneal injury. The hypoxic response is mediated partly through HIF1α and the mTOR inhibitor rapamycin blocks the hypoxic-induced angiogenic effects but does not affect the direct TGFß-mediated fibrosis and angiogenesis.


Assuntos
Neovascularização Patológica/patologia , Peritônio/irrigação sanguínea , Sirolimo/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Animais , Hipóxia Celular/efeitos dos fármacos , Células Cultivadas , Epitélio/efeitos dos fármacos , Epitélio/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Peritoneal/genética , Fibrose Peritoneal/patologia , Peritônio/patologia , Ratos , Proteína Smad3/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Am J Pathol ; 180(3): 940-951, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22203053

RESUMO

The mechanism of proteinuria in many common kidney diseases involves glomerular hemodynamic effects and local expression of angiogenic, fibrogenic, and vasoactive factors. Transforming growth factor (TGF)-ß has been associated with many diseases involving proteinuria and renal fibrosis. TGF-ß has been shown to induce podocyte dedifferentiation in vitro, but its in vivo effects on the glomerular filtration barrier are not well described. In this study, we used an adenovirus vector to transfer active TGF-ß1 to the glomeruli of rat kidneys. Transient TGF-ß1 overexpression induced significant proteinuria, podocyte foot process effacement, nephrin down-regulation, and nephrinuria. The expression of synaptopodin was also significantly down-regulated by TGF-ß1. Increased glomerular expression of Snail, suggestive of an in vivo dedifferentiation process, was associated with a loss of podocyte epithelial markers. The expression of angiopoietin-1 and angiopoietin-2 was significantly increased in TGF-ß1-transfected glomeruli, and TGF-ß1 increased the expression of the angiopoietin receptor, Tie2, in podocyte cell culture. TGF-ß1 down-regulated nephrin and synaptopodin expression in podocytes in cell culture; this effect was reversed by the blockade of both angiopoietin and Tie2 activities. These findings suggest that locally produced TGF-ß1 can cause podocyte dedifferentiation marked by a loss of synaptopodin, nephrin, and foot process effacement, partly regulated by angiopoietins. This process represents a novel pathway that may explain proteinuria in a variety of common renal diseases.


Assuntos
Proteinúria/etiologia , Fator de Crescimento Transformador beta1/fisiologia , Actinas/metabolismo , Adenoviridae , Angiopoietina-1/metabolismo , Angiopoietina-2/metabolismo , Animais , Desdiferenciação Celular , Células Cultivadas , Regulação para Baixo , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Barreira de Filtração Glomerular/metabolismo , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana/urina , Podócitos/metabolismo , Podócitos/patologia , Proteinúria/patologia , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição da Família Snail , Sinaptofisina/metabolismo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
11.
Am J Physiol Renal Physiol ; 300(2): F488-98, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21048020

RESUMO

Peritubular vascular changes and hypoxia after glomerular injury may explain subsequent tubulointerstitial injury and fibrosis. Several studies suggested that the expected tubulointerstitial angiogenic response is actively suppressed in this setting. The mechanism of this aberrant response has not been clearly identified. We used a common model of glomerular injury in rats to assess vascular changes and to identify potential factors associated with this aberrant response. Anti-Thy1.1 antibody administration (1 or 4 weekly doses) led to a dose-dependent renal damage characterized by elevated urea and tubulointerstitial fibrosis as assessed by Picro-Sirius Red staining. We quantified peritubular capillaries using CD31 and CD34 immunohistochemistry and showed that tubular angiogenic dysregulation was associated with peritubular capillary rarefaction. Using laser capture microdissection, we demonstrated an early induction of fibrogenic and angiogenic factors in the glomeruli and a subsequent dysregulated angiogenic response in the tubulointerstitial compartment. Proximal tubules of anti-Thy1.1-treated animals had increased pigment epithelial-derived factor (PEDF) expression by immunohistochemistry. Protein taken by laser capture microdissection also showed that PEDF was upregulated. Temporally associated with PEDF expression was a transient downregulation of tubular hypoxia-inducible factor (HIF)1α. In a human proximal tubular cell culture, we show that PEDF downregulates HIF1α protein and gene expression in cells exposed to 1% oxygen. In anti-Thy1.1 glomerulonephritis, there is aberrent tubular angiogenesis associated with glomerular injury and tubulointersititial fibrosis. We showed that PEDF may be involved by downregulating HIF1α. Further work is needed to elucidate the mechanism of PEDF upregulation and action in the tubules.


Assuntos
Glomerulonefrite/fisiopatologia , Túbulos Renais Proximais/fisiopatologia , Neovascularização Patológica/fisiopatologia , Animais , Antígenos CD34/análise , Antígenos CD34/imunologia , Linhagem Celular , Regulação para Baixo , Proteínas do Olho/metabolismo , Feminino , Glomerulonefrite/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Falência Renal Crônica/fisiopatologia , Túbulos Renais Proximais/irrigação sanguínea , Túbulos Renais Proximais/patologia , Fatores de Crescimento Neural/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Ratos , Ratos Sprague-Dawley , Serpinas/metabolismo , Antígenos Thy-1/imunologia , Regulação para Cima , Ureia/sangue , Fator A de Crescimento do Endotélio Vascular/análise , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Int J Nephrol ; 2010: 831243, 2010 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21188241

RESUMO

Objectives. The primary objective of this study was to determine the relationship between waist-to-hip ratio (WHR), cardiovascular (CV) events, and mortality in peritoneal dialysis (PD) patients. A secondary objective was to investigate the association between abdominal obesity and systemic inflammatory markers. Methods. This is a prospective study of 22 prevalent PD patients. WHR was measured at baseline. C-reactive protein (CRP), tumour necrosis factor-α (TNF-α), and interleukin-6 (IL-6) were measured. Main outcomes were first CV event and death from all causes. Survival analysis was used to examine the relationship between anthropomorphic measures and clinical outcomes. Results. Mean follow-up period was 3.1 years. In Kaplan-Meier analysis, survival was lower in those with higher WHR (P = .002). In Cox regression, WHR independently predicted mortality and first CV event after adjustment for known ischemic heart disease (hazard ratio [HR] 1.17, confidence interval [CI] 1.05-1.30 for death; HR 1.13, CI 1.01-1.26 for CV event). WHR correlated with serum TNF-α (r = 0.45; P = .05). Conclusion. The results of this study suggest WHR may be a risk factor for increased CV events and mortality in PD patients. Abdominal obesity is also associated with inflammatory markers. Larger studies are warranted to confirm these findings.

13.
Matrix Biol ; 29(2): 97-106, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19896531

RESUMO

Platelet derived growth factor (PDGF) is involved in wound healing in various organ systems. Its potential role in the context of peritoneal injury following long-term peritoneal dialysis is unclear. We used an adenovirus expressing the B chain of PDGF (AdPDGF-B) to assess its effect on pro-fibrotic pathways in the peritoneal membrane. To assess the transforming growth factor (TGF) beta independent effects of PDGF, we over-expressed PDGF-B in the peritoneum of either wild-type mice (Smad3+/+) or those with a deletion of the TGFbeta signaling protein Smad3 (Smad3(-/-)). PDGF-B induced sustained angiogenesis in both Smad3+/+ and Smad3(-/-) mice. Despite increased collagen gene expression, collagen accumulation was transient and fibrogenesis was associated with induction of collagenase activity. We observed epithelial to mesenchymal transition (EMT) involving the peritoneal mesothelial cells, as shown by increased SNAIL and decreased E-Cadherin expression with evidence of mesothelial cells expressing both epithelial and mesenchymal markers. Unlike TGFbeta-induced EMT, PDGF-B exposure did not lead to mobilization of the mesothelial cells; they remained as a single monolayer throughout the observation period. This "non-invasive" EMT phenomenon is a novel finding and may have implications concerning the role of EMT in peritoneal fibrosis and injury to other organ systems. The observed effects were similar in Smad3(-/-) and Smad3+/+ animals, suggesting that the PDGF-B effects were independent of TGFbeta or Smad signaling.


Assuntos
Epitélio/fisiologia , Mesoderma/fisiologia , Peritônio/citologia , Proteínas Proto-Oncogênicas c-sis/metabolismo , Proteína Smad3/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Diferenciação Celular , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Fibrose/patologia , Gelatinases/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Mesoderma/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Fisiológica/fisiologia , Peritônio/metabolismo , Peritônio/patologia , Fenótipo , Proteínas Proto-Oncogênicas c-sis/genética , Proteína Smad3/genética , Fator de Crescimento Transformador beta/metabolismo
14.
Kidney Int ; 77(4): 319-28, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19956083

RESUMO

Transition of peritoneal mesothelial cells to a mesenchymal phenotype plays an integral role in the angiogenic and fibrotic changes seen in the peritoneum of patients receiving long-term peritoneal dialysis. While signaling by transforming growth factor (TGF)-beta through Smad proteins likely causes these changes, it is possible that non-Smad pathways may also play a role. Here, we found that Smad3-deficient mice were protected from peritoneal fibrosis and angiogenesis caused by adenovirus-mediated gene transfer of active TGF-beta1 to mesothelial cells; however, mesothelial transition occurred in this setting, suggesting involvement of non-Smad mechanisms. The phosphatidyl inositol 3 kinase (PI3K) target, Akt, was upregulated in both Smad-deficient and wild-type mice after exposure to TGF-beta1. In vivo inhibition of the mammalian target of rapamycin (mTOR) by rapamycin completely abrogated the transition response in Smad3-deficient but not in wild-type mice. Rapamycin blocked nuclear localization of beta-catenin independent of glycogen synthase kinase 3beta activity. Further, in Smad3-deficient mice rapamycin reduced the expression of alpha-smooth muscle actin, which is an epithelial-to-mesenchymal transition-associated gene. Hence, we conclude that TGF-beta1 causes peritoneal injury through Smad-dependent and Smad-independent pathways; the latter involves redundant mechanisms inhibited by rapamycin, suggesting that suppression of both pathways may be necessary to abrogate mesothelial transition.


Assuntos
Fibrose Peritoneal/etiologia , Peritônio/patologia , Proteína Smad3/fisiologia , Animais , Diferenciação Celular , Células Epiteliais , Mesoderma/citologia , Camundongos , Transdução de Sinais
15.
Perit Dial Int ; 29(5): 508-16, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19776043

RESUMO

BACKGROUND: Encapsulating peritoneal sclerosis (EPS) is a rare complication of peritoneal dialysis. The causes of EPS are not well defined and are likely multifactorial. A suitable animal model would facilitate research into the pathophysiology and treatment of EPS. METHODS: We developed a helper-dependent adenovirus that expresses both green fluorescent protein (GFP) and active transforming growth factor-beta (TGF-beta1; HDAdTGF-beta1). Mice were administered HDAdTGF-beta1 via intraperitoneal injection and the response was compared with mice administered either first-generation adenovirus expressing TGF-beta1 (AdTGF-beta1) or control adenovirus (AdGFP). RESULTS: HDAdTGF-beta1-treated mice continued to express the GFP reporter transgene to day 74, the end of the observation period. Transgene expression lasted less than 28 days in the animals treated with first-generation adenoviruses. Animals treated with first-generation AdTGF-beta1 demonstrated submesothelial thickening and angiogenesis at day 7, with almost complete resolution by day 28. The HDAdTGF-beta1-treated mice demonstrated progressive peritoneal fibrosis with adhesion formation and encapsulation of bowels. Weight gain was significantly reduced in animals treated with HDAdTGF-beta1 compared to both the control-treated animals and the AdTGF-beta1-treated animals. Inflammation was not a major component of the fibroproliferative response. CONCLUSIONS: Peritoneal administration of a first-generation AdTGF-beta1 leads to transient gene expression, resulting in a resolving fibrotic response and histology similar to that seen in simple peritoneal sclerosis. Prolonged TGF-beta1 expression induced by the helper-dependent HDAdTGF-beta1 led to changes in peritoneal morphology resembling EPS. This suggests that TGF-beta1 may be a contributing factor in both simple peritoneal sclerosis and EPS. This model will be useful for elucidation of the mechanism of EPS and evaluation of potential treatment.


Assuntos
Adenoviridae/genética , Expressão Gênica , Vetores Genéticos , Vírus Auxiliares/genética , Fibrose Peritoneal/genética , Peritônio/metabolismo , Fator de Crescimento Transformador beta1/genética , Animais , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Diálise Peritoneal/efeitos adversos , Fibrose Peritoneal/etiologia , Fibrose Peritoneal/patologia , Peritônio/patologia
16.
Blood Purif ; 28(4): 354-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19729905

RESUMO

BACKGROUND/AIM: Volume expansion in peritoneal dialysis (PD) patients is associated with left ventricular hypertrophy. The link between inflammation and hypervolemia has not been extensively studied. The aim of this study was to determine if an association exists between hypervolemia and markers of inflammation in PD patients. METHODS: In this cross-sectional study of 22 prevalent PD patients, volume was determined by bioelectrical impedance analysis. Serum and peritoneal effluent interleukin-6 (IL-6) and peritoneal transforming growth factor (TGF)-beta(1) were measured. A fast peritoneal equilibration test determined peritoneal transport status. RESULTS: Bioimpedance-derived measures of hypervolemia correlated with peritoneal effluent IL-6 and TGF-beta(1). Peritoneal IL-6 was also associated with high peritoneal transport status. CONCLUSIONS: Markers of inflammation and fibrosis (peritoneal IL-6 and TGF-beta(1)) are associated with markers of hypervolemia.


Assuntos
Volume Sanguíneo , Fibrose/diagnóstico , Inflamação/diagnóstico , Diálise Peritoneal , Insuficiência Renal Crônica/patologia , Biomarcadores/análise , Estudos Transversais , Fibrose/etiologia , Humanos , Inflamação/etiologia , Interleucina-6/análise , Pessoa de Meia-Idade , Peritônio/química , Peritônio/patologia , Fator de Crescimento Transformador beta1/análise
17.
Am J Respir Crit Care Med ; 179(7): 588-94, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19151190

RESUMO

RATIONALE: The clinical management of idiopathic pulmonary fibrosis (IPF) remains a major challenge due to lack of effective drug therapy or accurate indicators for disease progression. Fibrocytes are circulating mesenchymal cell progenitors that are involved in tissue repair and fibrosis. OBJECTIVES: To test the hypothesis that assay of these cells may provide a biomarker for activity and progression of IPF. METHODS: Fibrocytes were defined as cells positive for CD45 and collagen-1 by flow cytometry and quantified in patients with stable IPF and during acute exacerbation of the disease. We investigated the clinical and prognostic value of fibrocyte counts by comparison with standard clinical parameters and survival. We used healthy age-matched volunteers and patients with acute respiratory distress syndrome as control subjects. MEASUREMENTS AND MAIN RESULTS: Fibrocytes were significantly elevated in patients with stable IPF (n = 51), with a further increase during acute disease exacerbation (n = 7; P < 0.001 vs. control subjects). Patients with acute respiratory distress syndrome (n = 10) were not different from healthy control subjects or stable patients with IPF. Fibrocyte numbers were not correlated with lung function or radiologic severity scores, but they were an independent predictor of early mortality. The mean survival of patients with fibrocytes higher than 5% of total blood leukocytes was 7.5 months compared with 27 months for patients with less than 5% (P < 0.0001). CONCLUSIONS: Fibrocytes are an indicator for disease activity of IPF and might be useful as a clinical marker for disease progression. This study suggests that quantification of circulating fibrocytes may allow prediction of early mortality in patients with IPF.


Assuntos
Fibrose Pulmonar Idiopática/sangue , Células-Tronco Mesenquimais/metabolismo , Idoso , Biomarcadores/sangue , Estudos de Casos e Controles , Contagem de Células , Feminino , Citometria de Fluxo , Humanos , Fibrose Pulmonar Idiopática/fisiopatologia , Antígenos Comuns de Leucócito , Masculino , Células-Tronco Mesenquimais/imunologia , Pessoa de Meia-Idade , Prognóstico , Modelos de Riscos Proporcionais , Estudos Prospectivos , Análise de Sobrevida
18.
Nephrol Dial Transplant ; 24(2): 448-57, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18775895

RESUMO

BACKGROUND: Morphological changes associated with long-term peritoneal dialysis (PD) include increased vascular surface area due to angiogenesis, submesothelial fibrosis and epithelial mesenchymal transition. Platelet-derived growth factor (PDGF) has been associated with all of these phenomena, and is a prototypical 'response to injury' growth factor. METHODS: Rats received an intraperitoneal injection of adenoviral vector expressing PDGF-B. At sacrifice, we analysed the structure and function of the peritoneal membrane. Gene expression in the peritoneal tissue was assessed for changes suggestive of epithelial mesenchymal transition. RESULTS: Over-expression of PDGF in the rat peritoneum led to significant angiogenesis, cellular proliferation and submesothelial thickening. Although PDGF induced expression of transforming growth factor beta, there was a lack of activation of this growth factor, and we believe that this explains the lack of significant collagen accumulation observed by a hydroxyproline assay. Despite evidence of angiogenesis and subsequent increased solute transport, we observed only a transient, non-significant impact on ultrafiltration function. This suggests that increased vascular surface area is necessary, but not sufficient, to produce ultrafiltration dysfunction. There was no evidence of epithelial mesenchymal transition observed either in regulation of associated genes such as Snail or E-Cadherin or in the lack of dual-labelled epithelial and mesenchymal cells on immunofluorescence. Mesothelial cells exposed to PDGF-B demonstrated increased collagen gene expression. CONCLUSIONS: PDGF-B induced angiogenesis without fibrosis in the peritoneum. The lack of significant ultrafiltration dysfunction and epithelial mesenchymal transition, as observed in patients on PD, suggests that PDGF-B may play a role, but is not the integral component, in response to peritoneal injury.


Assuntos
Peritônio/patologia , Peritônio/fisiopatologia , Proteínas Proto-Oncogênicas c-sis/fisiologia , Adenoviridae/genética , Animais , Colágeno/genética , Colágeno Tipo I , Epitélio/patologia , Expressão Gênica , Genes sis , Vetores Genéticos , Humanos , Mesoderma/patologia , Neovascularização Patológica , Diálise Peritoneal/efeitos adversos , Peritônio/irrigação sanguínea , Inibidor 1 de Ativador de Plasminogênio/genética , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas c-sis/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
19.
Exp Eye Res ; 88(2): 323-30, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18809398

RESUMO

Transforming growth factor beta (TGFbeta) has been known to play a role in anterior subcapsular cataract (ASC) formation and posterior capsule opacification (PCO), both of which are fibrotic pathologies of the lens. Several models have been utilized to study ASC formation, including the TGFbeta1 transgenic mouse model and the ex-vivo rat lens model. A distinct characteristic of ASC development within these models includes the formation of isolated fibrotic plaques or opacities which form beneath the lens capsule. A hallmark feature of ASC formation is the epithelial to mesenchymal transition (EMT) of lens epithelial cells (LECs) into myofibroblasts. Recently, the matrix metalloproteinases (MMPs) have been implicated in the formation of these cataracts through their involvement in EMT. In the present study, we sought to further investigate the role of MMPs in subcapsular cataract formation in a time course manner, through the examination of gene expression and morphological changes which occur during this process. RT-QPCR and immunohistochemical analysis was carried out on lenses treated with TGFbeta for a period of 2, 4 and 6 days. Laser capture microdissection (LCM) was utilized to specifically isolate cells within the plaque region and cells from the adjacent epithelium in lenses treated for a 6 day period. Multilayering of LECs was observed as early as day 2, which preceded the presence of alpha smooth muscle actin (alpha-SMA) immunoreactivity that was evident following 4 days of treatment with TGFbeta. A slight reduction in E-cadherin mRNA was detected at day 2, although this was not significant until the day 4 time point. Importantly, our results also indicate an early induction of MMP-9 mRNA following 2 days of TGFbeta treatment, whereas MMP-2 was found to be upregulated at the later 4 day time point. Further experiments using FHL 124 cells show an induction in MMP-2 protein levels following treatment with recombinant MMP-9. Together these findings suggest an upstream role for MMP-9 in ASC formation.


Assuntos
Catarata/enzimologia , Cápsula do Cristalino/enzimologia , Metaloproteinases da Matriz/genética , RNA Mensageiro/análise , Actinas/análise , Actinas/metabolismo , Animais , Caderinas/análise , Caderinas/metabolismo , Linhagem Celular , Proliferação de Células , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Humanos , Imuno-Histoquímica , Masculino , Metaloproteinase 2 da Matriz/análise , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/análise , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/farmacologia , Metaloproteinases da Matriz/análise , Microdissecção , Modelos Animais , Ratos , Ratos Wistar , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Fator de Crescimento Transformador beta/farmacologia
20.
Perit Dial Int ; 28(5): 497-504, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18708543

RESUMO

OBJECTIVE: To analyze gene expression of localized peritoneal tissue structures in a rodent model of peritoneal fibrosis. METHODS: Female Sprague Dawley rats were treated with an intraperitoneal injection of an adenovirus expressing active transforming growth factor-beta or control adenovirus. Four and 7 days after infection, animals were sacrificed and frozen sections of parietal peritoneum were subjected to immunofluorescence-aided laser capture microdissection in order to isolate vascular, mesothelial, and submesothelial structures. RNA was extracted from microdissected tissue and gene expression was analyzed by quantitative reverse-transcript polymerase chain reaction. We analyzed genes involved in angiogenesis, epithelial-to-mesenchymal transdifferentiation, and fibrosis. Vascular endothelial growth factor and alpha-smooth muscle actin expression was analyzed with immunohistochemistry of formalin-fixed tissue. RESULTS: Transforming growth factor-beta(1) induced expression of Snail and alpha-smooth muscle actin genes in the peritoneal mesothelium. This same cell population also demonstrated increased gene expression of vascular endothelial growth factor. The distribution of this growth factor was confirmed by immunohistochemistry. The fibrogenic growth factor, connective tissue growth factor, was also strongly induced in the peritoneal mesothelium. CONCLUSIONS: Using immunofluorescence-aided laser capture microdissection, we were able to study gene expression in subcompartments of the peritoneal tissue. We demonstrated that mesothelial cells exhibiting mesenchymal transdifferentiation are associated with increased expression of genes associated with fibrosis and angiogenesis.


Assuntos
Transdiferenciação Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Citometria de Varredura a Laser , Peritônio/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Actinas/metabolismo , Angiopoietina-2/metabolismo , Animais , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Feminino , Expressão Gênica , Microdissecção , Peritônio/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição da Família Snail , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA