Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Am J Physiol Renal Physiol ; 322(6): F597-F610, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35379003

RESUMEN

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.


Asunto(s)
Ácido Fólico , Receptor de Anafilatoxina C5a , Animales , Cicatriz , Fibrosis , Ácido Fólico/farmacología , Proteínas Fluorescentes Verdes , Riñón/patología , Ratones , Ratones Noqueados , Células Mieloides/patología , Receptor de Anafilatoxina C5a/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas
2.
Am J Physiol Renal Physiol ; 317(5): F1293-F1304, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31509012

RESUMEN

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.


Asunto(s)
Complemento C1r/metabolismo , Enfermedades Renales/enzimología , Animales , Línea Celular , Complemento C1r/genética , Complemento C1s/genética , Complemento C1s/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Ácido Fólico/farmacología , Regulación Enzimológica de la Expresión Génica , Humanos , Inflamación , Riñón/citología , Enfermedades Renales/genética , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo
3.
Am J Physiol Renal Physiol ; 312(3): F516-F532, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28052876

RESUMEN

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.


Asunto(s)
Activación de Complemento , Complemento C1q/metabolismo , Complemento C3/metabolismo , Túbulos Renales/metabolismo , Macrófagos/metabolismo , Pericitos/metabolismo , Insuficiencia Renal Crónica/metabolismo , Animales , Comunicación Celular , Complemento C1q/deficiencia , Complemento C1q/genética , Complemento C1q/inmunología , Complemento C3/deficiencia , Complemento C3/genética , Complemento C3/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteínas de la Matriz Extracelular/metabolismo , Fibrosis , Ácido Fólico , Genotipo , Mediadores de Inflamación/metabolismo , Túbulos Renales/inmunología , Túbulos Renales/patología , Antígenos Comunes de Leucocito/metabolismo , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Pericitos/inmunología , Pericitos/patología , Fenotipo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/inmunología , Insuficiencia Renal Crónica/patología , Factores de Tiempo , Obstrucción Ureteral/complicaciones , Vía de Señalización Wnt , Proteína Wnt3A/metabolismo
4.
Am J Pathol ; 185(5): 1198-200, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25747533

RESUMEN

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.


Asunto(s)
Células Endoteliales/metabolismo , Receptor IGF Tipo 1/metabolismo , Insuficiencia Renal Crónica/metabolismo , Animales , Humanos , Masculino
5.
J Am Soc Nephrol ; 26(4): 817-29, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25535303

RESUMEN

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.


Asunto(s)
Transdiferenciación Celular , Endotelio/metabolismo , Riñón/patología , Insuficiencia Renal Crónica/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Endoglina , Endotelio/patología , Fibrosis , Ácido Fólico , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones Transgénicos , Neovascularización Fisiológica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Insuficiencia Renal Crónica/patología , Proteínas Smad/metabolismo , Obstrucción Ureteral
6.
Kidney Int ; 87(1): 95-108, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24940804

RESUMEN

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.


Asunto(s)
Proteína HMGB1/metabolismo , Sirtuina 1/fisiología , Acetilación , Animales , Células Cultivadas , Células Endoteliales , Humanos , Ratones
7.
Prostaglandins Other Lipid Mediat ; 120: 17-20, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25936481

RESUMEN

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.


Asunto(s)
Comunicación Celular , Células/citología , Células/metabolismo , Animales , Células/patología , Humanos , Isquemia/patología , Macrófagos/citología , Células Madre Mesenquimatosas/citología , Sepsis/patología
8.
J Am Soc Nephrol ; 25(2): 276-91, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24136919

RESUMEN

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.


Asunto(s)
Metaloproteinasa 14 de la Matriz/fisiología , Nefroesclerosis/enzimología , Sirtuina 1/deficiencia , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Animales , Senescencia Celular , Concanavalina A/farmacología , Regulación hacia Abajo , Endotelio Vascular/fisiopatología , Exones/genética , Matriz Extracelular/metabolismo , Fibrosis , Ácido Fólico/toxicidad , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Riñón/enzimología , Riñón/patología , Riñón/fisiopatología , Fallo Renal Crónico/inducido químicamente , Fallo Renal Crónico/metabolismo , Masculino , Metaloproteinasa 14 de la Matriz/genética , Ratones , Ratones Mutantes , Ratones Transgénicos , Neovascularización Fisiológica , Nefroesclerosis/genética , Nefroesclerosis/patología , Regeneración , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Sirtuina 1/fisiología , Vasodilatación
9.
Am J Physiol Heart Circ Physiol ; 307(12): H1691-704, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25239805

RESUMEN

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.


Asunto(s)
Cardiomegalia/metabolismo , Diástole , Endotelio Vascular/metabolismo , Neovascularización Fisiológica , Sirtuina 1/metabolismo , Función Ventricular , Factores de Edad , Animales , Vasos Coronarios/crecimiento & desarrollo , Vasos Coronarios/metabolismo , Vasos Coronarios/fisiología , Fibrosis/metabolismo , Ventrículos Cardíacos/crecimiento & desarrollo , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Sirtuina 1/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Am J Pathol ; 180(3): 973-983, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22234173

RESUMEN

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.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Catepsinas/farmacología , Senescencia Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Sirtuina 1/metabolismo , Células Madre/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Arginina/análogos & derivados , Arginina/farmacología , Autofagia/efectos de los fármacos , Azoles/farmacología , Permeabilidad de la Membrana Celular/fisiología , Células Cultivadas , Colágeno Tipo I/farmacología , Regulación hacia Abajo , Endotelio Vascular , Peróxido de Hidrógeno/farmacología , Isoindoles , Lisosomas/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Compuestos de Organoselenio/farmacología , Proteínas Recombinantes , Factores de Riesgo , Sirtuina 1/deficiencia , Estrés Fisiológico/efectos de los fármacos
11.
Br J Pharmacol ; 178(14): 2880-2891, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33555070

RESUMEN

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.


Asunto(s)
Activación de Complemento , Enfermedades Renales , Animales , Fibrosis , Riñón , Macrófagos , Ratones
12.
J Am Soc Nephrol ; 20(10): 2127-37, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19679673

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas del Citoesqueleto/fisiología , Podocitos/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal , Proteína Smad2/fisiología , Proteína smad3/fisiología , Animales , Apoptosis , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Riñón/patología , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/fisiología , Proteinuria/etiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Esclerosis
13.
Stem Cells Transl Med ; 6(3): 992-1005, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28297566

RESUMEN

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.


Asunto(s)
Microambiente Celular , Riñón/patología , Células 3T3 , Animales , Microambiente Celular/efectos de los fármacos , Quimiocinas/metabolismo , Receptor gp130 de Citocinas/metabolismo , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/efectos de los fármacos , Células Progenitoras Endoteliales/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibrosis , Interleucina-6/farmacología , Factor Inhibidor de Leucemia/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miofibroblastos/citología , Miofibroblastos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Factores de Transcripción/metabolismo , Obstrucción Ureteral/patología
14.
Int J Radiat Oncol Biol Phys ; 65(2): 517-27, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16690434

RESUMEN

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.


Asunto(s)
Ácido 4-Aminobenzoico/uso terapéutico , Suplementos Dietéticos , Melanocitos/efectos de los fármacos , Melanocitos/efectos de la radiación , Melanoma/radioterapia , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Animales , Apoptosis/efectos de la radiación , Western Blotting , Línea Celular Tumoral , Proliferación Celular , Embrión de Pollo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Melanocitos/metabolismo , Melanoma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa
15.
Biochim Biophys Acta ; 1573(2): 109-20, 2002 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-12399020

RESUMEN

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.


Asunto(s)
Antioxidantes/farmacología , Daño del ADN , Hidroxilaminas/farmacología , Óxidos de Nitrógeno/farmacología , Protectores contra Radiación/farmacología , Animales , Línea Celular , Supervivencia Celular , Cobre , Óxidos N-Cíclicos/farmacología , ADN/análisis , ADN/efectos de los fármacos , ADN/efectos de la radiación , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Electroforesis/métodos , Peróxido de Hidrógeno , Oxidación-Reducción , Fenantrolinas , Plásmidos , Marcadores de Spin , Triacetonamina-N-Oxil/farmacología
16.
Free Radic Biol Med ; 34(1): 93-102, 2003 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12498984

RESUMEN

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.


Asunto(s)
Anticarcinógenos/farmacología , Antioxidantes/farmacología , Óxidos N-Cíclicos/farmacología , Animales , Anticarcinógenos/química , Antioxidantes/química , Peso Corporal/efectos de los fármacos , Óxidos N-Cíclicos/química , Femenino , Masculino , Ratones , Ratones Endogámicos C3H , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Peso Molecular , Proteínas/metabolismo , Marcadores de Spin
17.
Ann N Y Acad Sci ; 962: 195-206, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12076975

RESUMEN

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.


Asunto(s)
Óxido Nítrico/química , Especies de Nitrógeno Reactivo/química , Especies Reactivas de Oxígeno/química , Antioxidantes/química , Antioxidantes/metabolismo , Óxido Nítrico/metabolismo , Oxidación-Reducción , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo
18.
PLoS One ; 8(9): e75572, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24086574

RESUMEN

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.


Asunto(s)
Apoptosis/genética , Regulación hacia Abajo/genética , MicroARNs/genética , MicroARNs/metabolismo , Podocitos/metabolismo , Proteína Smad2/genética , Factor de Crecimiento Transformador beta/genética , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Línea Celular , Humanos , Etiquetado Corte-Fin in Situ , Ratones , Ratones Transgénicos , Transducción de Señal/genética , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Antioxid Redox Signal ; 19(3): 211-30, 2013 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-23088293

RESUMEN

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.


Asunto(s)
Lesión Renal Aguda/metabolismo , Autofagia , Células Endoteliales/metabolismo , Estrés Oxidativo , Peroxisomas/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Lesión Renal Aguda/inducido químicamente , Acil-CoA Oxidasa/metabolismo , Albuminuria , Animales , Catalasa/metabolismo , Citocinas/sangre , Modelos Animales de Enfermedad , Activación Enzimática , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Mediadores de Inflamación/sangre , Lipopolisacáridos/efectos adversos , Ratones , Modelos Biológicos , Oxidación-Reducción , Transporte de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción TFIIH , Factores de Transcripción/metabolismo
20.
PLoS One ; 7(6): e39406, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761782

RESUMEN

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.


Asunto(s)
Células Endoteliales/metabolismo , Homeostasis/fisiología , Proteínas de la Membrana/metabolismo , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Movimiento Celular/fisiología , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Glomérulos Renales/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Nefrectomía , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA