Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Am J Physiol Renal Physiol ; 322(6): F597-F610, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35379003

RESUMO

We have previously reported that increased expression and activation of kidney cell complement components play an important role in the pathogenesis of renal scarring. Here, we used floxed green fluorescent protein (GFP)-C5a receptor 1 (C5aR1) knockin mice (GFP-C5ar1fl/fl) and the model of folic acid (FA)-induced kidney injury to define the cell types and potential mechanisms by which increased C5aR1 activation leads to fibrosis. Using flow cytometry and confocal microscopy, we identified macrophages as the major interstitial cell type showing increased expression of C5aR1 in FA-treated mice. C5ar1fl/fl.Lyz2Cre+/- mice, in which C5aR1 has been specifically deleted in lysozyme M-expressing myeloid cells, experienced reduced fibrosis compared with control C5ar1fl/fl mice. Examination of C5aR1-expressing macrophage transcriptomes by gene set enrichment analysis demonstrated that these cells were enriched in pathways corresponding to the complement cascade, collagen formation, and the NABA matrisome, strongly pointing to their critical roles in tissue repair/scarring. Since C5aR1 was also detected in a small population of platelet-derived growth factor receptor-ß+ GFP+ cells, we developed C5ar1fl/fl.Foxd1Cre+/- mice, in which C5aR1 is deleted specifically in pericytes, and found reduced FA-induced fibrosis. Primary cell cultures of platelet-derived growth factor receptor-ß+ pericytes isolated from FA-treated C5ar1fl/fl.Foxd1Cre+/- mice showed reduced secretion of several cytokines, including IL-6 and macrophage inflammatory protein-2, compared with pericytes isolated from FA-treated control GFP-C5ar1fl/fl mice. Collectively, these data imply that C5a/C5aR1 axis activation primarily in interstitial cells contributes to the development of renal fibrosis.NEW & NOTEWORTHY This study used novel green fluorescent protein C5a receptor 1 floxed mice and the model of folic acid-mediated kidney fibrosis to demonstrate the pathogenic role of increased expression of this complement receptor on macrophages.


Assuntos
Ácido Fólico , Receptor da Anafilatoxina C5a , Animais , Cicatriz , Fibrose , Ácido Fólico/farmacologia , Proteínas de Fluorescência Verde , Rim/patologia , Camundongos , Camundongos Knockout , Células Mieloides/patologia , Receptor da Anafilatoxina C5a/genética , Receptores do Fator de Crescimento Derivado de Plaquetas
2.
Am J Physiol Renal Physiol ; 317(5): F1293-F1304, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31509012

RESUMO

We have previously reported that complement activation precedes the development of kidney fibrosis; however, little is known about the cellular mechanisms involved in this transition. We hypothesized that increased expression of C1 complex protease C1r, the initiator of complement activation, contributes to tubulointerstitial fibrosis and tested this idea in mice with global deletion of C1r. Although expression of C1r in untreated wild-type (WT) mice was higher in the liver compared with kidney tissue, administration of folic acid (FA) led to upregulation of C1r mRNA and protein levels only in kidney tissue. Immunohistochemistry and in situ hybridization experiments localized increased expression of C1r and C1s proteases to renal tubular epithelial cells. C1r-null mice had reduced acute tubular injury and inflammation measured 2 days after FA administration compared with WT mice. C1r deletion reduced expression of C1s, C3 fragment formation, and organ fibrosis measured 14 days after FA administration. Differential gene expression performed in kidney tissue demonstrated that C1r-null mice had reduced expression of genes associated with the acute phase response, complement, proliferation of connective tissue cells (e.g., platelet-derived growth factor receptor-ß), and reduced expression of genes associated with inflammation compared with FA-treated WT mice. In vitro experiments in renal epithelial cells demonstrated that C1s expression is dependent on increased C1r expression and that interferon-γ induces the expression of these two proteases. We conclude that increased expression of C1 complex proteases is associated with increased tissue inflammation and complement C3 formation and represents an important pathogenic mechanism leading to FA-mediated tubulointerstitial fibrosis.


Assuntos
Complemento C1r/metabolismo , Nefropatias/enzimologia , Animais , Linhagem Celular , Complemento C1r/genética , Complemento C1s/genética , Complemento C1s/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Ácido Fólico/farmacologia , Regulação Enzimológica da Expressão Gênica , Humanos , Inflamação , Rim/citologia , Nefropatias/genética , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
3.
Am J Physiol Renal Physiol ; 312(3): F516-F532, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28052876

RESUMO

We have examined the pathogenic role of increased complement expression and activation during kidney fibrosis. Here, we show that PDGFRß-positive pericytes isolated from mice subjected to obstructive or folic acid injury secrete C1q. This was associated with increased production of proinflammatory cytokines, extracellular matrix components, collagens, and increased Wnt3a-mediated activation of Wnt/ß-catenin signaling, which are hallmarks of myofibroblast activation. Real-time PCR, immunoblots, immunohistochemistry, and flow cytometry analysis performed in whole kidney tissue confirmed increased expression of C1q, C1r, and C1s as well as complement activation, which is measured as increased synthesis of C3 fragments predominantly in the interstitial compartment. Flow studies localized increased C1q expression to PDGFRß-positive pericytes as well as to CD45-positive cells. Although deletion of C1qA did not prevent kidney fibrosis, global deletion of C3 reduced macrophage infiltration, reduced synthesis of C3 fragments, and reduced fibrosis. Clodronate mediated depletion of CD11bF4/80 high macrophages in UUO mice also reduced complement gene expression and reduced fibrosis. Our studies demonstrate local synthesis of complement by both PDGFRß-positive pericytes and CD45-positive cells in kidney fibrosis. Inhibition of complement activation represents a novel therapeutic target to ameliorate fibrosis and progression of chronic kidney disease.


Assuntos
Ativação do Complemento , Complemento C1q/metabolismo , Complemento C3/metabolismo , Túbulos Renais/metabolismo , Macrófagos/metabolismo , Pericitos/metabolismo , Insuficiência Renal Crônica/metabolismo , Animais , Comunicação Celular , Complemento C1q/deficiência , Complemento C1q/genética , Complemento C1q/imunologia , Complemento C3/deficiência , Complemento C3/genética , Complemento C3/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Proteínas da Matriz Extracelular/metabolismo , Fibrose , Ácido Fólico , Genótipo , Mediadores da Inflamação/metabolismo , Túbulos Renais/imunologia , Túbulos Renais/patologia , Antígenos Comuns de Leucócito/metabolismo , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pericitos/imunologia , Pericitos/patologia , Fenótipo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/imunologia , Insuficiência Renal Crônica/patologia , Fatores de Tempo , Obstrução Ureteral/complicações , Via de Sinalização Wnt , Proteína Wnt3A/metabolismo
4.
Am J Pathol ; 185(5): 1198-200, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25747533

RESUMO

This commentary highlights the article by Liang et al that describes a critical role for insulin-like growth factor 1 receptor in the progression of chronic kidney disease.


Assuntos
Células Endoteliais/metabolismo , Receptor IGF Tipo 1/metabolismo , Insuficiência Renal Crônica/metabolismo , Animais , Humanos , Masculino
5.
J Am Soc Nephrol ; 26(4): 817-29, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25535303

RESUMO

Excessive TGF-ß signaling in epithelial cells, pericytes, or fibroblasts has been implicated in CKD. This list has recently been joined by endothelial cells (ECs) undergoing mesenchymal transition. Although several studies focused on the effects of ablating epithelial or fibroblast TGF-ß signaling on development of fibrosis, there is a lack of information on ablating TGF-ß signaling in the endothelium because this ablation causes embryonic lethality. We generated endothelium-specific heterozygous TGF-ß receptor knockout (TßRII(endo+/-)) mice to explore whether curtailed TGF-ß signaling significantly modifies nephrosclerosis. These mice developed normally, but showed enhanced angiogenic potential compared with TßRII(endo+/+) mice under basal conditions. After induction of folic acid nephropathy or unilateral ureteral obstruction, TßRII(endo+/-) mice exhibited less tubulointerstitial fibrosis, enhanced preservation of renal microvasculature, improvement in renal blood flow, and less tissue hypoxia than TßRII(endo+/+) counterparts. In addition, partial deletion of TßRII in the endothelium reduced endothelial-to-mesenchymal transition (EndoMT). TGF-ß-induced canonical Smad2 signaling was reduced in TßRII(+/-) ECs; however, activin receptor-like kinase 1 (ALK1)-mediated Smad1/5 phosphorylation in TßRII(+/-) ECs remained unaffected. Furthermore, the S-endoglin/L-endoglin mRNA expression ratio was significantly lower in TßRII(+/-) ECs compared with TßRII(+/+) ECs. These observations support the hypothesis that EndoMT contributes to renal fibrosis and curtailing endothelial TGF-ß signals favors Smad1/5 proangiogenic programs and dictates increased angiogenic responses. Our data implicate endothelial TGF-ß signaling and EndoMT in regulating angiogenic and fibrotic responses to injury.


Assuntos
Transdiferenciação Celular , Endotélio/metabolismo , Rim/patologia , Insuficiência Renal Crônica/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Endoglina , Endotélio/patologia , Fibrose , Ácido Fólico , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos Transgênicos , Neovascularização Fisiológica , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Insuficiência Renal Crônica/patologia , Proteínas Smad/metabolismo , Obstrução Ureteral
6.
Kidney Int ; 87(1): 95-108, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24940804

RESUMO

High-mobility group box 1 (HMGB1) undergoes acetylation, nuclear-to-cytoplasmic translocation, and release from stressed kidneys, unleashing a signaling cascade of events leading to systemic inflammation. Here, we tested whether the deacetylase activity of Sirtuin1 (SIRT1) participates in regulating nuclear retention of HMGB1 to ultimately modulate damage signaling initiated by HMGB1 secretion during stress. When immunoprecipitated acetylated HMGB1 was incubated with SIRT1, HMGB1 acetylation decreased by 57%. Proteomic analysis showed that SIRT1 deacetylates HMGB1 at four lysine residues (55, 88, 90, and 177) within the proinflammatory and nuclear localization signal domains of HMGB1. Genetic ablation or pharmacological inhibition of SIRT1 in endothelial cells increased HMGB1 acetylation and translocation. In vivo, deletion of SIRT1 reduced nuclear HMGB1 while increasing its acetylation and release into circulation during basal and ischemic conditions, causing increased renal damage. Conversely, resveratrol pretreatment led to decreased HMGB1 acetylation, its nuclear retention, decreased systemic release, and reduced tubular damage. Thus, a vicious cycle is set into motion in which the inflammation-induced repression of SIRT1 disables deacetylation of HMGB1, facilitates its nuclear-to-cytoplasmic translocation, and systemic release, thereby maintaining inflammation.


Assuntos
Proteína HMGB1/metabolismo , Sirtuína 1/fisiologia , Acetilação , Animais , Células Cultivadas , Células Endoteliais , Humanos , Camundongos
7.
Prostaglandins Other Lipid Mediat ; 120: 17-20, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25936481

RESUMO

We are witnessing the emergence of a novel type of biological regulation, namely, the communication between cells via their secreted substances, the secretome. This brief overview is based on the available published data and our own experience. We discuss three vignettes illustrating the importance of communication via the secretome: (1) the secretome of stem cells and its effects in sepsis and systemic inflammatory response; (2) the profibrotic secretomes partially responsible for development of fibrotic complications; and (3) the contribution of senescence-associated secretory products to the propagation of the senescence phenotype. Considering the richness of secretomes of different cells under diverse conditions, it becomes imperative to gain insights into their individual components in an attempt to harness cell secretomes for therapeutic purposes.


Assuntos
Comunicação Celular , Células/citologia , Células/metabolismo , Animais , Células/patologia , Humanos , Isquemia/patologia , Macrófagos/citologia , Células-Tronco Mesenquimais/citologia , Sepse/patologia
8.
J Am Soc Nephrol ; 25(2): 276-91, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24136919

RESUMO

Sirtuin 1 (SIRT1) depletion in vascular endothelial cells mediates endothelial dysfunction and premature senescence in diverse cardiovascular and renal diseases. However, the molecular mechanisms underlying these pathologic effects remain unclear. Here, we examined the phenotype of a mouse model of vascular senescence created by genetically ablating exon 4 of Sirt1 in endothelial cells (Sirt1(endo-/-)). Under basal conditions, Sirt1(endo-/-) mice showed impaired endothelium-dependent vasorelaxation and angiogenesis, and fibrosis occurred spontaneously at low levels at an early age. In contrast, induction of nephrotoxic stress (acute and chronic folic acid-induced nephropathy) in Sirt1(endo-/-) mice resulted in robust acute renal functional deterioration followed by an exaggerated fibrotic response compared with control animals. Additional studies identified matrix metalloproteinase-14 (MMP-14) as a target of SIRT1. In the kidneys of Sirt1(endo-/-) mice, impaired angiogenesis, reduced matrilytic activity, and retention of the profibrotic cleavage substrates tissue transglutaminase and endoglin accompanied MMP-14 suppression. Furthermore, restoration of MMP-14 expression in SIRT1-depeleted mice improved angiogenic and matrilytic functions of the endothelium, prevented renal dysfunction, and attenuated nephrosclerosis. Our findings establish a novel mechanistic molecular link between endothelial SIRT1 depletion, downregulation of MMP-14, and the development of nephrosclerosis.


Assuntos
Metaloproteinase 14 da Matriz/fisiologia , Nefroesclerose/enzimologia , Sirtuína 1/deficiência , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/metabolismo , Animais , Senescência Celular , Concanavalina A/farmacologia , Regulação para Baixo , Endotélio Vascular/fisiopatologia , Éxons/genética , Matriz Extracelular/metabolismo , Fibrose , Ácido Fólico/toxicidade , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Rim/enzimologia , Rim/patologia , Rim/fisiopatologia , Falência Renal Crônica/induzido quimicamente , Falência Renal Crônica/metabolismo , Masculino , Metaloproteinase 14 da Matriz/genética , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Neovascularização Fisiológica , Nefroesclerose/genética , Nefroesclerose/patologia , Regeneração , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/genética , Sirtuína 1/fisiologia , Vasodilatação
9.
Am J Physiol Heart Circ Physiol ; 307(12): H1691-704, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25239805

RESUMO

Discordant myocardial growth and angiogenesis can explain left ventricular (LV) hypertrophy progressing toward heart failure with aging. Sirtuin 1 expression declines with age; therefore we explored the role played by angiogenesis and Sirtuin 1 in the development of cardiomyopathy. We compared the cardiac function of 10- to 15-wk-old (wo), 30-40 wo, and 61-70 wo endothelial Sirtuin 1-deleted (Sirt1(endo-/-)) mice and their corresponding knockout controls (Sirt1(Flox/Flox)). After 30-40 wk, Sirt1(endo-/-) animals exhibited diastolic dysfunction (DD), decreased mRNA expression of Serca2a in the LV, and decreased capillary density compared with control animals despite a similar VEGFa mRNA expression. However, LV fibrosis and hypoxia-inducible factor (HIF)1α expression were not different. The creation of a transverse aortic constriction (TAC) provoked more severe DD and LV fibrosis in Sirt1(endo-/-) compared with control TAC animals. Although the VEGFa mRNA expression was not different and the protein expression of HIF1α was higher in the Sirt1(endo-/-) TAC animals, capillary density remained reduced. In cultured endothelial cells administration of Sirtuin 1 inhibitor decreased mRNA expression of VEGF receptors FLT 1 and FLK 1. Ex vivo capillary sprouting from aortic explants showed impaired angiogenic response to VEGF in the Sirt1(endo-/-) mice. In conclusion, the data demonstrate 1) a defect in angiogenesis preceding development of DD; 2) dispensability of endothelial Sirtuin 1 under unstressed conditions and during normal aging; and 3) impaired angiogenic adaptation and aggravated DD in Sirt1(endo-/-) mice challenged with LV overload.


Assuntos
Cardiomegalia/metabolismo , Diástole , Endotélio Vascular/metabolismo , Neovascularização Fisiológica , Sirtuína 1/metabolismo , Função Ventricular , Fatores Etários , Animais , Vasos Coronários/crescimento & desenvolvimento , Vasos Coronários/metabolismo , Vasos Coronários/fisiologia , Fibrose/metabolismo , Ventrículos do Coração/crescimento & desenvolvimento , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Sirtuína 1/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Am J Pathol ; 180(3): 973-983, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22234173

RESUMO

Stress-induced premature senescence (SIPS) of endothelial cells (ECs) has emerged as a contributor to global EC dysfunction. One of the cellular abnormalities mechanistically linked to SIPS is lysosomal dysfunction. In this study, we examined the impact of a range of cardiovascular risk factors on the expression of sirtuin 1 (SIRT1), SIPS, and apoptosis, and we documented the role of SIRT1 in reduced EC and endothelial progenitor cell (EPC) viability. These findings were confirmed in mice with selective endothelial SIRT1 knockout. The effects of stressors could be partially mimicked by inducing lysosomal membrane permeabilization or inhibiting autophagy, and were reversed by a cathepsin inhibitor. We provide evidence that SIRT1 is an important substrate of cysteine cathepsins B, S, and L. An antioxidant/peroxynitrite scavenger, ebselen, prevented stress-induced SIRT1 depletion and subversion of autophagy by mitigating lysosomal dysfunction. In conclusion, our data advance the concept of "stem cell aging" by establishing the critical role of lysosomal dysfunction in the development of SIPS through the cathepsin-induced proteolytic cleavage of SIRT1, a mechanism linking cell stress to apoptosis and SIPS. Ebselen potently protects lysosomal membrane integrity, preventing cathepsin-induced cleavage of SIRT 1 in EPCs and blunting SIPS and apoptotic cell death induced by relevant cardiovascular stressors. The proposed mechanism of SIRT1 depletion in stress has all of the attributes of being a paradigm of SIPS of EPCs.


Assuntos
Doenças Cardiovasculares/etiologia , Catepsinas/farmacologia , Senescência Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Sirtuína 1/metabolismo , Células-Tronco/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Arginina/análogos & derivados , Arginina/farmacologia , Autofagia/efeitos dos fármacos , Azóis/farmacologia , Permeabilidade da Membrana Celular/fisiologia , Células Cultivadas , Colágeno Tipo I/farmacologia , Regulação para Baixo , Endotélio Vascular , Peróxido de Hidrogênio/farmacologia , Isoindóis , Lisossomos/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Compostos Organosselênicos/farmacologia , Proteínas Recombinantes , Fatores de Risco , Sirtuína 1/deficiência , Estresse Fisiológico/efeitos dos fármacos
11.
Br J Pharmacol ; 178(14): 2880-2891, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33555070

RESUMO

Increased expression of complement C1r, C1s and C3 in kidney cells plays an important role in the pathogenesis of kidney fibrosis. Our studies suggest that activation of complement in kidney cells with increased generation of C3 and its fragments occurs by activation of classical and alternative pathways. Single nuclei RNA sequencing studies in kidney tissue from unilateral ureteral obstruction mice show that increased synthesis of complement C3 and C5 occurs primarily in renal tubular epithelial cells (proximal and distal), while increased expression of complement receptors C3ar1 and C5ar1 occurs in interstitial cells including immune cells like monocytes/macrophages suggesting compartmentalization of complement components during kidney injury. Although global deletion of C3 and macrophage ablation prevent inflammation and reduced kidney tissue scarring, the development of mice with cell-specific deletion of complement components and their regulators could bring further insights into the mechanisms by which intracellular complement activation leads to fibrosis and progressive kidney disease. LINKED ARTICLES: This article is part of a themed issue on Canonical and non-canonical functions of the complement system in health and disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.14/issuetoc.


Assuntos
Ativação do Complemento , Nefropatias , Animais , Fibrose , Rim , Macrófagos , Camundongos
12.
J Am Soc Nephrol ; 20(10): 2127-37, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19679673

RESUMO

TGF-beta regulates differentiation, growth, and apoptosis of podocytes and mediates podocyte depletion in glomerulosclerosis. TGF-beta promotes proapoptotic signaling mediated by Smad3 but also activates prosurvival pathways such as phosphoinositide-3 kinase (PI3K)/AKT; the latter requires the CD2-associated adaptor protein (CD2AP) in podocytes. Whether the opposing activities mediated by Smad proteins and CD2AP involve molecular cross-talk is unknown. Here, we report that CD2AP-dependent early activation of the antiapoptotic PI3K/AKT pathway does not require TGF-beta receptor-regulated Smad2 and Smad3. We found that the C-terminal region of CD2AP interacts directly with the cytoplasmic tail of the TGF-beta receptor type I (TbetaRI) in a kinase-dependent manner and that the interaction between the TbetaRI and the p85 subunit of PI3K requires CD2AP. Consistent with the proapoptotic function of Smad signaling, Smad2/3-deficient podocytes were hyperproliferative and resistant to TGF-beta-induced growth inhibition and apoptosis. In contrast, CD2AP-deficient cells were hypoproliferative and hypersensitive to TGF-beta-induced apoptosis. In vivo, to determine the effects of reduced Smad3 or CD2AP gene dosage on podocyte apoptosis and proteinuria characteristic of TGF-beta1 transgenic mice, we generated TGF-beta1 transgenic mice deficient for Smad3 or heterozygous for CD2AP. Smad3 deficiency ameliorated podocyte apoptosis, and CD2AP heterozygosity increased both podocyte apoptosis and proteinuria. These data define distinct canonical (Smad) and noncanonical (CD2AP/PI3K/AKT) pathways that arise from direct, independent interactions with the TbetaRI and that mediate opposing signals for podocyte death or survival.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas do Citoesqueleto/fisiologia , Podócitos/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Transdução de Sinais , Proteína Smad2/fisiologia , Proteína Smad3/fisiologia , Animais , Apoptose , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Rim/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/fisiologia , Proteinúria/etiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptor do Fator de Crescimento Transformador beta Tipo I , Esclerose
13.
Stem Cells Transl Med ; 6(3): 992-1005, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28297566

RESUMO

Accumulation of myofibroblasts is a hallmark of renal fibrosis. A significant proportion of myofibroblasts has been reported to originate via endothelial-mesenchymal transition. We initially hypothesized that exposing myofibroblasts to the extract of endothelial progenitor cells (EPCs) could reverse this transition. Indeed, in vitro treatment of transforming growth factor-ß1 (TGF-ß1)-activated fibroblasts with EPC extract prevented expression of α-smooth muscle actin (α-SMA); however, it did not enhance expression of endothelial markers. In two distinct models of renal fibrosis-unilateral ureteral obstruction and chronic phase of folic acid-induced nephropathy-subcapsular injection of EPC extract to the kidney prevented and reversed accumulation of α-SMA-positive myofibroblasts and reduced fibrosis. Screening the composition of EPC extract for cytokines revealed that it is enriched in leukemia inhibitory factor (LIF) and vascular endothelial growth factor. Only LIF was capable of reducing fibroblast-to-myofibroblast transition of TGF-ß1-activated fibroblasts. In vivo subcapsular administration of LIF reduced the number of myofibroblasts and improved the density of peritubular capillaries; however, it did not reduce the degree of fibrosis. A receptor-independent ligand for the gp130/STAT3 pathway, hyper-interleukin-6 (hyper-IL-6), not only induced a robust downstream increase in pluripotency factors Nanog and c-Myc but also exhibited a powerful antifibrotic effect. In conclusion, EPC extract prevented and reversed fibroblast-to-myofibroblast transition and renal fibrosis. The component of EPC extract, LIF, was capable of preventing development of the contractile phenotype of activated fibroblasts but did not eliminate TGF-ß1-induced collagen synthesis in cultured fibroblasts and models of renal fibrosis, whereas a receptor-independent gp130/STAT3 agonist, hyper-IL-6, prevented fibrosis. In summary, these studies, through the evolution from EPC extract to LIF and then to hyper-IL-6, demonstrate the instructive role of microenvironmental cues and may provide in the future a facile strategy to prevent and reverse renal fibrosis. Stem Cells Translational Medicine 2017;6:992-1005.


Assuntos
Microambiente Celular , Rim/patologia , Células 3T3 , Animais , Microambiente Celular/efeitos dos fármacos , Quimiocinas/metabolismo , Receptor gp130 de Citocina/metabolismo , Células Progenitoras Endoteliais/citologia , Células Progenitoras Endoteliais/efeitos dos fármacos , Células Progenitoras Endoteliais/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibrose , Interleucina-6/farmacologia , Fator Inibidor de Leucemia/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miofibroblastos/citologia , Miofibroblastos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Fatores de Transcrição/metabolismo , Obstrução Ureteral/patologia
14.
Int J Radiat Oncol Biol Phys ; 65(2): 517-27, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16690434

RESUMO

PURPOSE: To determine whether para-aminobenzoic acid (PABA) alters the sensitivity of tumor cells to ionizing radiation in vitro and in vivo. METHODS AND MATERIALS: Cellular proliferation was assessed by WST-1 assays. The effects of PABA and radiation on tumor growth were examined with chick embryo and murine models. Real-time reverse transcriptase-polymerase chain reaction and Western blotting were used to quantify p21CIP1 and CDC25A levels. RESULTS: Para-aminobenzoic acid enhanced (by 50%) the growth inhibitory activity of radiation on B16F10 cells, whereas it had no effect on melanocytes. Para-aminobenzoic acid enhanced (50-80%) the antitumor activity of radiation on B16F10 and 4T1 tumors in vivo. The combination of PABA and radiation therapy increased tumor apoptosis. Treatment of tumor cells with PABA increased expression of CDC25A and decreased levels of p21CIP1. CONCLUSIONS: Our findings suggest that PABA might represent a compound capable of enhancing the antitumor activity of ionizing radiation by a mechanism involving altered expression of proteins known to regulate cell cycle arrest.


Assuntos
Ácido 4-Aminobenzoico/uso terapêutico , Suplementos Nutricionais , Melanócitos/efeitos dos fármacos , Melanócitos/efeitos da radiação , Melanoma/radioterapia , Radiossensibilizantes/uso terapêutico , Animais , Apoptose/efeitos da radiação , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Embrião de Galinha , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Melanócitos/metabolismo , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase
15.
Biochim Biophys Acta ; 1573(2): 109-20, 2002 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-12399020

RESUMO

Modulation of radiation- and metal ion-catalyzed oxidative-induced damage using plasmid DNA, genomic DNA, and cell survival, by three nitroxides and their corresponding hydroxylamines, were examined. The antioxidant property of each compound was independently determined by reacting supercoiled DNA with copper II/1,10-phenanthroline complex fueled by the products of hypoxanthine/xanthine oxidase (HX/XO) and noting the protective effect as assessed by agarose gel electrophoresis. The nitroxides and their corresponding hydroxylamines protected approximately to the same degree (33-47% relaxed form) when compared to 76.7% relaxed form in the absence of protectors. Likewise, protection by both the nitroxide and corresponding hydroxylamine were observed for Chinese hamster V79 cells exposed to hydrogen peroxide. In contrast, when plasmid DNA damage was induced by ionizing radiation (100 Gy), only nitroxides (10 mM) provide protection (32.4-38.5% relaxed form) when compared to radiation alone or in the presence of hydroxylamines (10 mM) (79.8% relaxed form). Nitroxide protection was concentration dependent. Radiation cell survival studies and DNA double-strand break (DBS) assessment (pulse field electrophoresis) showed that only the nitroxide protected or prevented damage, respectively. Collectively, the results show that nitroxides and hydroxylamines protect equally against the damage mediated by oxidants generated by the metal ion-catalyzed Haber-Weiss reaction, but only nitroxides protect against radiation damage, suggesting that nitroxides may more readily react with intermediate radical species produced by radiation than hydroxylamines.


Assuntos
Antioxidantes/farmacologia , Dano ao DNA , Hidroxilaminas/farmacologia , Óxidos de Nitrogênio/farmacologia , Protetores contra Radiação/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular , Cobre , Óxidos N-Cíclicos/farmacologia , DNA/análise , DNA/efeitos dos fármacos , DNA/efeitos da radiação , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Eletroforese/métodos , Peróxido de Hidrogênio , Oxirredução , Fenantrolinas , Plasmídeos , Marcadores de Spin , Triacetonamina-N-Oxil/farmacologia
16.
Free Radic Biol Med ; 34(1): 93-102, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12498984

RESUMO

Stable free radical nitroxides are potent antioxidants possessing superoxide dismutase- and catalase-mimetic activity that protect cells and animals against a variety of oxidative insults. Tempol, as a representative nitroxide, was evaluated for its influence on weight maintenance and spontaneous tumor incidence in C3H mice. Tempol administered in either the drinking water or food did not show any untoward effects and prevented animals from becoming obese. Tempol-treated animals' leptin levels were reduced. Long-term treatment with Tempol significantly decreased tumorigenesis when compared to controls (10 vs. 40%, respectively). Selected tissues from Tempol-treated animals exhibited elevated levels of mitochrondrial uncoupling protein-2 (UCP-2) and HSP70. The present data suggest that nitroxides upregulate UCP-2, obviate weight gain, and decrease age-related spontaneous tumor incidence. As a class, nitroxides may provide overall health benefits by contributing to decreased obesity and tumor incidence.


Assuntos
Anticarcinógenos/farmacologia , Antioxidantes/farmacologia , Óxidos N-Cíclicos/farmacologia , Animais , Anticarcinógenos/química , Antioxidantes/química , Peso Corporal/efeitos dos fármacos , Óxidos N-Cíclicos/química , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C3H , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Peso Molecular , Proteínas/metabolismo , Marcadores de Spin
17.
Ann N Y Acad Sci ; 962: 195-206, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12076975

RESUMO

Nitric oxide (nitrogen monoxide, NO) plays a veritable cornucopia of regulatory roles in normal physiology. In contrast, NO has also been implicated in the etiology and sequela of numerous neurodegenerative diseases that involve reactive oxygen species (ROS) and nitrogen oxide species (RNOS). In this setting, NO is often viewed solely as pathogenic; however, the chemistry of NO can also be a significant factor in lessening injury mediated by both ROS and RNOS. The relationship between NO and oxidation, nitrosation, and nitration reactions is summarized. The salient factors that determine whether NO promotes, abates, or interconnects these chemistries are emphasized. From this perspective of NO chemistry, the type, magnitude, location, and duration of either ROS or RNOS reactions may be predicted.


Assuntos
Óxido Nítrico/química , Espécies Reativas de Nitrogênio/química , Espécies Reativas de Oxigênio/química , Antioxidantes/química , Antioxidantes/metabolismo , Óxido Nítrico/metabolismo , Oxirredução , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo
18.
PLoS One ; 8(9): e75572, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086574

RESUMO

Transforming growth factors beta (TGF-ß) are multi-functional cytokines capable of inducing apoptosis in epithelial cells, including glomerular podocytes. We and others have previously shown that podocyte-selective genetic deletion of the microRNA (miR)-processing enzyme, Dicer, caused glomerulosclerosis that was associated with podocyte apoptosis, and the miR-30 family was implicated in the process. Here, we report that apoptosis-associated genes were highly enriched among the predicted targets of miR-30 when compared with randomly selected miRs (26% vs. 4.5 ± 2.1%) or with the known TGF-ß-regulated miR-192 (6%), miR-216a (5.1%), and miR-217 (0%). miR-30 family members were abundantly expressed in podocytes in normal mice but were downregulated in albumin/TGF-ß transgenic mice with podocyte apoptosis and glomerulosclerosis. In vitro, TGF-ß downregulated miR-30s in wildtype and Smad3-deficient, but not Smad2- or Smad2/Smad3-deficient, podocytes. The TGF-ß-induced activation of caspase 3 and an increase in TUNEL-positive nuclei were significantly inhibited by the lentivirus-mediated overexpression of miR-30d, but not by a scrambled control miR, in podocytes. TGF-ß stimulated the phosphorylation of pro-apoptotic p53 in podocytes with lentiviral expression of a scrambled miR, but not in podocytes expressing miR-30d. In contrast, miR-30d had no effect on the phosphorylation of pro-apoptotic p38 MAP kinase induced by TGF-ß. Thus, we report that Smad2-dependent inhibition of miR-30s in podocytes is required for the activation of p53 and the induction of apoptosis by TGF-ß. These results demonstrate a novel functional role for miR-30 in podocyte survival and indicate that the loss of miR-30 survival signaling is a novel and specific mechanism of TGF-ß-induced podocyte apoptosis during glomerulosclerosis. We propose the therapeutic replacement of miR-30 as a novel strategy to prevent the podocyte apoptosis that is characteristic of progressive glomerular diseases.


Assuntos
Apoptose/genética , Regulação para Baixo/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Podócitos/metabolismo , Proteína Smad2/genética , Fator de Crescimento Transformador beta/genética , Animais , Caspase 3/genética , Caspase 3/metabolismo , Linhagem Celular , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Transgênicos , Transdução de Sinais/genética , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Antioxid Redox Signal ; 19(3): 211-30, 2013 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-23088293

RESUMO

AIMS: We examined that (a) how the endotoxic stress affects peroxisomal function and autophagic degradation of peroxisomes-pexophagy, (b) how a superimposed dysfunction of lysosomes and pexophagy modifies responses to lipopolysaccharide (LPS), and (c) the mechanisms of peroxisomal contribution to renal injury. To accomplish this, we used lysosome-defective Lyst-mice in vivo and primary endothelial cells in vitro, and compared the responses with wild-type (WT) littermates. RESULTS: LPS induced pexophagic degradation, followed by proliferation of peroxisomes in WT mice, which was abolished in Lyst-mice. Lyst-mice exhibited impaired activation of catalase, which together with preserved hydrogen peroxide-generating ß-oxidation resulted in redox disequilibrium. LPS treatment induced a heightened inflammatory response, increased oxidative damage, and aggravated renal injury in Lyst-mice. Similarly, as in vivo, LPS-activated lysosomal (LYS) pexophagy and transiently repressed peroxisomes in vitro, supported by reduced peroxisomal density in the vicinity of lysosomes. Peroxisomal dynamics was also abolished in lysosome-defective cells, which accumulated peroxisomes with compromised functions and intraorganellar redox imbalance. INNOVATION: We demonstrated that pexophagy is a default response to endotoxic injury. However, when LYS dysfunction (a frequent companion of chronic diseases) is superimposed, recycling and functioning of peroxisomes are impaired, and an imbalance between hydrogen peroxide-generating ß-oxidation and hydrogen peroxide-detoxifying catalase ensues, which ultimately results in peroxisomal burnout. CONCLUSION: Our data strongly suggest that pexophagy, a cellular mechanism per se, is essential in functional maintenance of peroxisomes during LPS exposure. Inhibition of pexophagy results in accumulation of impaired peroxisomes, redox disequilibrium, and aggravated renal damage.


Assuntos
Injúria Renal Aguda/metabolismo , Autofagia , Células Endoteliais/metabolismo , Estresse Oxidativo , Peroxissomos/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Injúria Renal Aguda/induzido quimicamente , Acil-CoA Oxidase/metabolismo , Albuminúria , Animais , Catalase/metabolismo , Citocinas/sangue , Modelos Animais de Doenças , Ativação Enzimática , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Mediadores da Inflamação/sangue , Lipopolissacarídeos/efeitos adversos , Camundongos , Modelos Biológicos , Oxirredução , Transporte Proteico , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição TFIIH , Fatores de Transcrição/metabolismo
20.
PLoS One ; 7(6): e39406, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761782

RESUMO

BACKGROUND: BAMBI is a type I TGFß receptor antagonist, whose in vivo function remains unclear, as BAMBI(-/-) mice lack an obvious phenotype. METHODOLOGY/PRINCIPAL FINDINGS: Identifying BAMBI's functions requires identification of cell-specific expression of BAMBI. By immunohistology we found BAMBI expression restricted to endothelial cells and by electron microscopy BAMBI(-/-) mice showed prominent and swollen endothelial cells in myocardial and glomerular capillaries. In endothelial cells over-expression of BAMBI reduced, whereas knock-down enhanced capillary growth and migration in response to TGFß. In vivo angiogenesis was enhanced in matrigel implants and in glomerular hypertrophy after unilateral nephrectomy in BAMBI(-/-) compared to BAMBI(+/+) mice consistent with an endothelial phenotype for BAMBI(-/-) mice. BAMBI's mechanism of action in endothelial cells was examined by canonical and alternative TGFß signaling in HUVEC with over-expression or knock-down of BAMBI. BAMBI knockdown enhanced basal and TGFß stimulated SMAD1/5 and ERK1/2 phosphorylation, while over-expression prevented both. CONCLUSIONS/SIGNIFICANCE: Thus we provide a first description of a vascular phenotype for BAMBI(-/-) mice, and provide in vitro and in vivo evidence that BAMBI contributes to endothelial and vascular homeostasis. Further, we demonstrate that in endothelial cells BAMBI interferes with alternative TGFß signaling, most likely through the ALK 1 receptor, which may explain the phenotype observed in BAMBI(-/-) mice. This newly described role for BAMBI in regulating endothelial function has potential implications for understanding and treating vascular disease and tumor neo-angiogenesis.


Assuntos
Células Endoteliais/metabolismo , Homeostase/fisiologia , Proteínas de Membrana/metabolismo , Neovascularização Fisiológica/fisiologia , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Movimento Celular/fisiologia , Endotélio Vascular/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Glomérulos Renais/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Nefrectomia , Fosforilação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA