Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 81(1): 244, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38814462

RESUMEN

Four-and-a-half LIM domains protein 2 (FHL2) is an adaptor protein that may interact with hypoxia inducible factor 1α (HIF-1α) or ß-catenin, two pivotal protective signaling in acute kidney injury (AKI). However, little is known about the regulation and function of FHL2 during AKI. We found that FHL2 was induced in renal tubular cells in patients with acute tubular necrosis and mice model of ischemia-reperfusion injury (IRI). In cultured renal proximal tubular cells (PTCs), hypoxia induced FHL2 expression and promoted the binding of HIF-1 to FHL2 promoter. Compared with control littermates, mice with PTC-specific deletion of FHL2 gene displayed worse renal function, more severe morphologic lesion, more tubular cell death and less cell proliferation, accompanying by downregulation of AQP1 and Na, K-ATPase after IRI. Consistently, loss of FHL2 in PTCs restricted activation of HIF-1 and ß-catenin signaling simultaneously, leading to attenuation of glycolysis, upregulation of apoptosis-related proteins and downregulation of proliferation-related proteins during IRI. In vitro, knockdown of FHL2 suppressed hypoxia-induced activation of HIF-1α and ß-catenin signaling pathways. Overexpression of FHL2 induced physical interactions between FHL2 and HIF-1α, ß-catenin, GSK-3ß or p300, and the combination of these interactions favored the stabilization and nuclear translocation of HIF-1α and ß-catenin, enhancing their mediated gene transcription. Collectively, these findings identify FHL2 as a direct downstream target gene of HIF-1 signaling and demonstrate that FHL2 could play a critical role in protecting against ischemic AKI by promoting the activation of HIF-1 and ß-catenin signaling through the interactions with its multiple protein partners.


Asunto(s)
Lesión Renal Aguda , Túbulos Renales Proximales , Proteínas con Homeodominio LIM , Proteínas Musculares , Daño por Reperfusión , Factores de Transcripción , beta Catenina , Animales , Proteínas con Homeodominio LIM/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Lesión Renal Aguda/genética , Humanos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/genética , Ratones , beta Catenina/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Masculino , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Transducción de Señal , Ratones Endogámicos C57BL , Ratones Noqueados , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Proliferación Celular , Apoptosis
2.
Am J Pathol ; 193(8): 1029-1045, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37236504

RESUMEN

IL-33, a member of the IL-1 family, acts as an alarmin in immune response. Epithelial-mesenchymal transition and transforming growth factor-ß (TGF-ß)­induced fibroblast activation are key events in the development of renal interstitial fibrosis. The current study found increased expression of IL-33 and interleukin-1 receptor-like 1 (IL1RL1, alias ST2), the receptor for IL-33, in human fibrotic renal tissues. In addition, IL-33­ or ST2-deficient mice showed significantly reduced levels of fibronectin, α-smooth muscle actin, and vimentin, and increased E-cadherin levels. In HK-2 cells, IL-33 promotes the phosphorylation of the TGF-ß receptor (TGF-ßR), Smad2, and Smad3, and the production of extracellular matrix (ECM), with reduced expression of E-cadherin. Blocking TGF-ßR signaling or suppressing ST2 expression impeded Smad2 and Smad3 phosphorylation, thereby reducing ECM production, suggesting that IL-33­induced ECM synthesis requires cooperation between the two pathways. Mechanistically, IL-33 treatment induced a proximate interaction between ST2 and TGF-ßRs, activating downstream Smad2 and Smad3 for ECM production in renal epithelial cells. Collectively, this study identified a novel and essential role for IL-33 in promoting TGF-ß signaling and ECM production in the development of renal fibrosis. Therefore, targeting IL-33/ST2 signaling may be an effective therapeutic strategy for renal fibrosis.


Asunto(s)
Interleucina-33 , Enfermedades Renales , Ratones , Humanos , Animales , Interleucina-33/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/uso terapéutico , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Enfermedades Renales/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteína smad3/metabolismo , Fibrosis , Cadherinas/metabolismo , Factores de Crecimiento Transformadores/metabolismo , Factores de Crecimiento Transformadores/farmacología , Factores de Crecimiento Transformadores/uso terapéutico , Factor de Crecimiento Transformador beta1/metabolismo , Transición Epitelial-Mesenquimal
3.
J Biol Chem ; 298(7): 102010, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35525270

RESUMEN

Follistatin (FS)-like 1 (FSTL1) is a member of the FS-SPARC (secreted protein, acidic and rich in cysteine) family of secreted and extracellular matrix proteins. The functions of FSTL1 have been studied in heart and lung injury as well as in wound healing; however, the role of FSTL1 in the kidney is largely unknown. Here, we show using single-cell RNA-Seq that Fstl1 was enriched in stromal cells in obstructed mouse kidneys. In addition, immunofluorescence demonstrated that FSTL1 expression was induced in fibroblasts during kidney fibrogenesis in mice and human patients. We demonstrate that FSTL1 overexpression increased renal fibrosis and activated the Wnt/ß-catenin signaling pathway, known to promote kidney fibrosis, but not the transforming growth factor ß (TGF-ß), Notch, Hedgehog, or Yes-associated protein (YAP) signaling pathways in obstructed mouse kidneys, whereas inhibition of FSTL1 lowered Wnt/ß-catenin signaling. Importantly, we show that FSTL1 interacted with Wnt ligands and the Frizzled (FZD) receptors but not the coreceptor lipoprotein receptor-related protein 6 (LRP6). Specifically, we found FSTL1 interacted with Wnt3a through its extracellular calcium-binding (EC) domain and von Willebrand factor type C-like (VWC) domain, and with FZD4 through its EC domain. Furthermore, we show that FSTL1 increased the association of Wnt3a with FZD4 and promoted Wnt/ß-catenin signaling and fibrogenesis. The EC domain interacting with both Wnt3a and FZD4 also enhanced Wnt3a signaling. Therefore, we conclude that FSTL1 is a novel extracellular enhancer of the Wnt/ß-catenin pathway.


Asunto(s)
Proteínas Relacionadas con la Folistatina , Receptores Frizzled , Riñón , Vía de Señalización Wnt , Animales , Proteínas Relacionadas con la Folistatina/genética , Proteínas Relacionadas con la Folistatina/metabolismo , Receptores Frizzled/metabolismo , Humanos , Riñón/metabolismo , Riñón/fisiopatología , Ligandos , Ratones , Proteína Wnt3A
4.
Kidney Int ; 102(2): 321-336, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35483524

RESUMEN

Energy metabolism is crucial in maintaining cellular homeostasis and adapting to stimuli for tubular cells. However, the underlying mechanisms remain largely unknown. Here, we report that PP2Acα was upregulated in damaged tubular cells from patients and animal models with acute or chronic kidney injury. Using in vitro and in vivo model, we demonstrated that PP2Acα induction in damaged tubular cells suppresses fatty acid oxidation and promotes glycolysis, leading to cell death and fibrosis. Mechanistically, we revealed that PP2Acα dephosphorylates ACC through interaction with B56δ, leading to the regulation of fatty acid oxidation. Furthermore, PP2Acα also dephosphorylates p-Glut1 (Thr478) and suppresses Trim21-mediated Glut1 ubiquitination and degradation, leading to the promotion of glucose intake and glycolysis. Thus, this study adds new insight into the tubular cell metabolic alterations in kidney diseases. PP2Acα may be a promising therapeutic target for kidney injury.


Asunto(s)
Glucólisis , Riñón , Animales , Ácidos Grasos , Transportador de Glucosa de Tipo 1 , Fosfoproteínas Fosfatasas
5.
FASEB J ; 35(7): e21706, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34160104

RESUMEN

Acute kidney injury (AKI) is a devastating condition with high morbidity and mortality rates. The pathological features of AKI are tubular injury, infiltration of inflammatory cells, and impaired vascular integrity. Pyruvate kinase is the final rate-limiting enzyme in the glycolysis pathway. We previously showed that pyruvate kinase M2 (PKM2) plays an important role in regulating the glycolytic reprogramming of fibroblasts in renal interstitial fibrosis. The present study aimed to determine the role of PKM2 in fibroblast activation during the pathogenesis of AKI. We found increased numbers of S100A4 positive cells expressing PKM2 in renal tissues from mice with AKI induced via folic acid or ischemia/reperfusion (I/R). The loss of PKM2 in fibroblasts impaired fibroblast proliferation and promoted tubular epithelial cell death including apoptosis, necroptosis, and ferroptosis. Mechanistically, fibroblasts produced less hepatocyte growth factor (HGF) in response to a loss of PKM2. Moreover, in two AKI mouse models, fibroblast-specific deletion of PKM2 blocked HGF signal activation and aggravated AKI after it was induced in mice via ischemia or folic acid. Fibroblast proliferation mediated by PKM2 elicits pro-survival signals that repress tubular cell death and may help to prevent AKI progression. Fibroblast activation mediated by PKM2 in AKI suggests that targeting PKM2 expression could be a novel strategy for treating AKI.


Asunto(s)
Lesión Renal Aguda/metabolismo , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Piruvato Quinasa/metabolismo , Animales , Apoptosis/fisiología , Muerte Celular/fisiología , Línea Celular , Modelos Animales de Enfermedad , Fibrosis/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Necroptosis/fisiología , Daño por Reperfusión/metabolismo , Transducción de Señal/fisiología
6.
Kidney Int ; 98(3): 686-698, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32739207

RESUMEN

Energy reprogramming to glycolysis is closely associated with the development of chronic kidney disease. As an important negative regulatory factor of the mammalian target of rapamycin complex 1 (mTORC1) signal, tuberous sclerosis complex 1 (Tsc1) is also a key regulatory point of glycolysis. Here, we investigated whether Tsc1 could mediate the progression of kidney interstitial fibrosis by regulating glycolysis in proximal tubular epithelial cells. We induced mTORC1 signal activation in tubular epithelial cells in kidneys with fibrosis via unilateral ureteral occlusion. This resulted in increased tubular epithelial cell proliferation and glycolytic enzyme upregulation. Prior incubation with rapamycin inhibited mTORC1 activation and abolished the enhanced glycolysis and tubular epithelial cell proliferation. Furthermore, knockdown of Tsc1 expression promoted glycolysis in the rat kidney epithelial cell line NRK-52E. Specific deletion of Tsc1 in the proximal tubules of mice resulted in enlarged kidneys characterized by a high proportion of proliferative tubular epithelial cells, dilated tubules with cyst formation, and a large area of interstitial fibrosis in conjunction with elevated glycolysis. Treatment of the mice with the glycolysis inhibitor 2-deoxyglucose notably ameliorated tubular epithelial cell proliferation, cystogenesis, and kidney fibrosis. Thus, our findings suggest that Tsc1-associated mTORC1 signaling mediates the progression of kidney interstitial fibrosis by regulating glycolysis in proximal tubular epithelial cells.


Asunto(s)
Esclerosis Tuberosa , Animales , Células Epiteliales , Fibrosis , Glucólisis , Riñón/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratas , Esclerosis Tuberosa/metabolismo , Esclerosis Tuberosa/patología , Proteína 1 del Complejo de la Esclerosis Tuberosa
7.
J Cell Sci ; 131(6)2018 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-29361523

RESUMEN

Renal fibrosis is a final common pathway of chronic kidney disease. Sustained activation of fibroblasts is considered to play a key role in perpetuating renal fibrosis but the driving force in the perpetuation stage is only partially understood. To date, some investigations have specifically identified overexpression of microRNA 21 (miR-21) in the progression of kidney fibrosis. Nevertheless, the precise role of miR-21 in fibroblast activation remains largely unknown. In this study, we found that miR-21 was significantly upregulated in activated fibroblasts and that it maintained itself at constant high levels by employing an auto-regulatory loop between miR-21, PDCD4 and AP-1. Persistently upregulated miR-21 suppressed protein expression of Smad7 and, eventually, enhanced the TGF-ß1/Smad pathway to promote fibroblast activation. More importantly, we found miR-21 sequestration with miR-21 antagomir or AP-1 inhibitors attenuated unilateral ureteral obstruction (UUO)-induced renal fibrosis. miR-21-knockout mice also suffered far less interstitial fibrosis in response to kidney injury. Altogether, these data suggest that miR-21 is a main driving force of fibroblast activation and keeps its high expression level by employing a double negative autoregulatory loop. Targeting this aberrantly activated feedback loop may provide new therapeutic strategy in treating fibrotic kidneys.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas de Unión al ARN/metabolismo , Insuficiencia Renal Crónica/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Fibroblastos/metabolismo , Fibrosis/genética , Fibrosis/patología , Humanos , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas de Unión al ARN/genética , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/patología , Transducción de Señal , Proteína smad7/genética , Proteína smad7/metabolismo , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba
8.
J Biol Chem ; 293(42): 16364-16375, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30154246

RESUMEN

Our previously published study demonstrated that mammalian target of rapamycin complex 2 (mTORC2) signaling mediates TGFß1-induced fibroblast activation. However, the underlying mechanisms for mTORC2 in stimulating fibroblast activation remain poorly understood. Here, we found that TGFß1 could stimulate mTORC2 and Yap/Taz activation in NRK-49F cells. Blocking either mTORC2 or Yap/Taz signaling diminished TGFß1-induced fibroblast activation. In addition, blockade of mTORC2 could down-regulate the expression of Yap/Taz, connective tissue growth factor (CTGF), and ankyrin repeat domain 1 (ANKRD1). Overexpression of constitutively active Taz (Taz-S89A) could restore fibroblast activation suppressed by PP242, an mTOR kinase inhibitor in NRK-49F cells. In mouse kidneys with unilateral ureter obstructive (UUO) nephropathy, both mTORC2 and Yap/Taz were activated in the interstitial myofibroblasts. Ablation of Rictor in fibroblasts/pericytes or blockade of mTOR signaling with PP242 attenuated Yap/Taz activation and UUO nephropathy in mice. Together, this study uncovers that targeting mTORC2 retards fibroblast activation and kidney fibrosis through suppressing Yap/Taz activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Fibroblastos/metabolismo , Enfermedades Renales/patología , Diana Mecanicista del Complejo 2 de la Rapamicina/fisiología , Fosfoproteínas/metabolismo , Factores de Transcripción/metabolismo , Aciltransferasas , Animales , Proteínas de Ciclo Celular , Línea Celular , Fibrosis , Ratones , Factor de Crecimiento Transformador beta1/farmacología , Proteínas Señalizadoras YAP
9.
J Biol Chem ; 293(50): 19290-19302, 2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30333225

RESUMEN

M2 macrophage polarization is known to underlie kidney fibrosis. We previously reported that most of the members of the Wnt family of signaling proteins are induced in fibrotic kidneys. Dysregulation of the signaling protein Wnt5a is associated with fibrosis, but little is known about the role of Wnt5a in regulating M2 macrophage activation that results in kidney fibrosis. Here, using murine Raw 264.7 cells and bone marrow-derived macrophages, we found that Wnt5a enhanced transforming growth factor ß1 (TGFß1)-induced macrophage M2 polarization as well as expression of the transcriptional regulators Yes-associated protein (Yap)/transcriptional coactivator with PDZ-binding motif (Taz). Verteporfin blockade of Yap/Taz inhibited both Wnt5a- and TGFß1-induced macrophage M2 polarization. In mouse models of kidney fibrosis, shRNA-mediated knockdown of Wnt5a expression diminished kidney fibrosis, macrophage Yap/Taz expression, and M2 polarization. Moreover, genetic ablation of Taz in macrophages attenuated kidney fibrosis and macrophage M2 polarization in mice. Collectively, these results indicate that Wnt5a promotes kidney fibrosis by stimulating Yap/Taz-mediated macrophage M2 polarization.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Riñón/patología , Macrófagos/citología , Fosfoproteínas/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Proteína Wnt-5a/metabolismo , Animales , Proteínas de Ciclo Celular , Regulación hacia Abajo , Fibrosis , Masculino , Ratones , Ratones Endogámicos C57BL , Transactivadores , Transcripción Genética , Proteínas Señalizadoras YAP
10.
J Biol Chem ; 293(28): 11119-11130, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-29794026

RESUMEN

Kidney fibrosis is a histological hallmark of chronic kidney disease and arises in large part through extracellular matrix deposition by activated fibroblasts. The signaling protein complex mTOR complex 2 (mTORC2) plays a critical role in fibroblast activation and kidney fibrosis. Protein kinase Cα (PKCα) is one of the major sub-pathways of mTORC2, but its role in fibroblast activation and kidney fibrosis remains to be determined. Here, we found that transforming growth factor ß1 (TGFß1) activates PKCα signaling in cultured NRK-49F cells in a time-dependent manner. Blocking PKCα signaling with the chemical inhibitor Go6976 or by transfection with PKCα siRNA largely reduced expression of the autophagy-associated protein lysosomal-associated membrane protein 2 (LAMP2) and also inhibited autophagosome-lysosome fusion and autophagic flux in the cells. Similarly to chloroquine, Go6976 treatment and PKCα siRNA transfection also markedly inhibited TGFß1-induced fibroblast activation. In murine fibrotic kidneys with unilateral ureteral obstruction (UUO) nephropathy, PKCα signaling is activated in the interstitial myofibroblasts. Go6976 administration largely blocked autophagic flux in fibroblasts in the fibrotic kidneys and attenuated the UUO nephropathy. Together, our findings suggest that blocking PKCα activity may retard autophagic flux and thereby prevent fibroblast activation and kidney fibrosis.


Asunto(s)
Autofagia , Fibroblastos/patología , Fibrosis/patología , Enfermedades Renales/patología , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Proteína Quinasa C-alfa/metabolismo , Obstrucción Ureteral/patología , Animales , Células Cultivadas , Fibroblastos/metabolismo , Fibrosis/metabolismo , Enfermedades Renales/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Masculino , Ratones , Proteína Quinasa C-alfa/genética , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/metabolismo
11.
J Am Soc Nephrol ; 29(1): 182-193, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29021383

RESUMEN

The Wnt/ß-catenin pathway is crucial in normal development and throughout life, but aberrant activation of this pathway has been linked to kidney fibrosis, although the mechanisms involved remain incompletely determined. Here, we investigated the role of Wnt/ß-catenin in regulating macrophage activation and the contribution thereof to kidney fibrosis. Treatment of macrophages with Wnt3a exacerbated IL-4- or TGFß1-induced macrophage alternative (M2) polarization and the phosphorylation and nuclear translocation of STAT3 in vitro Conversely, inhibition of Wnt/ß-catenin signaling prevented these IL-4- or TGFß1-induced processes. In a mouse model, induced deletion of ß-catenin in macrophages attenuated the fibrosis, macrophage accumulation, and M2 polarization observed in the kidneys of wild-type littermates after unilateral ureter obstruction. This study shows that activation of Wnt/ß-catenin signaling promotes kidney fibrosis by stimulating macrophage M2 polarization.


Asunto(s)
Riñón/patología , Activación de Macrófagos , Macrófagos/fisiología , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Línea Celular , Fibrosis , Interleucina-4/metabolismo , Masculino , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/complicaciones , Proteína Wnt3A/farmacología
12.
J Cell Mol Med ; 22(3): 1684-1695, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29193729

RESUMEN

ß-Catenin signalling plays an important role in regulating tubular epithelial-to-mesenchymal transition (EMT), an indispensable programme for driving renal fibrosis. As an adapter protein, four and a half LIM domain protein 2 (FHL2) acts as a coregulator of ß-catenin in several other cell types. To determine whether FHL2 affects ß-catenin signalling and thus is involved in tubular EMT, we examined its expression and function in the process of TGF-ß1-induced EMT. FHL2 mRNA and protein were induced by TGF-ß1 in rat tubular epithelial cells (NRK-52E), an effect that intracellular Smad signalling was required. Ectopic expression of FHL2 inhibited E-cadherin and enhanced α-smooth muscle actin (α-SMA) and fibronectin expression, whereas knockdown of FHL2 partially restored E-cadherin and reduced α-SMA and fibronectin induction stimulated by TGF-ß1. Overexpression of FHL2 increased ß-catenin dephosphorylation (Ser37/Thr41), nuclear translocation and ß-catenin-mediated transcription and up-regulated expression of ß-catenin target, EMT-related genes, such as Snail, Twist, vimentin, plasminogen activator inhibitor-1 and matrix metalloproteinase-7. Conversely, knockdown of FHL2 increased ß-catenin phosphorylation (Ser33/37/Thr41), decreased its nuclear translocation and inhibited ß-catenin-mediated transcription and target genes expression. TGF-ß1 induced a FHL2/ß-catenin interaction in NRK-52E cells, especially in the nuclei. In a mouse model of obstructive nephropathy, FHL2 mRNA and protein were induced in a time-dependent fashion, and the extent and pattern of renal ß-catenin activation were positively correlated with FHL2 induction. Collectively, this study suggests that FHL2, via modulating ß-catenin signalling, may implicate in regulation of TGF-ß1-mediated tubular EMT and could be a potential therapeutic target for fibrotic kidney disease.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Túbulos Renales Proximales/efectos de los fármacos , Proteínas con Homeodominio LIM/metabolismo , Proteínas Musculares/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , beta Catenina/metabolismo , Animales , Línea Celular , Transición Epitelial-Mesenquimal/genética , Expresión Génica/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Proteínas con Homeodominio LIM/genética , Masculino , Ratones , Proteínas Musculares/genética , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Interferencia de ARN , Ratas , Transducción de Señal/genética , Factores de Transcripción/genética , beta Catenina/genética
13.
J Pathol ; 242(4): 488-499, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28585302

RESUMEN

Mammalian target of rapamycin (mTOR) signalling controls many essential cellular functions. However, the role of Rictor/mTOR complex 2 (mTORC2) in regulating macrophage activation and kidney fibrosis remains largely unknown. We report here that Rictor/mTORC2 was activated in macrophages from the fibrotic kidneys of mice. Ablation of Rictor in macrophages reduced kidney fibrosis, inflammatory cell accumulation, macrophage proliferation and polarization after unilateral ureter obstruction or ischaemia/reperfusion injury. In bone marrow-derived macrophages (BMMs), deletion of Rictor or blockade of protein kinase Cα inhibited cell migration. Additionally, deletion of Rictor or blockade of Akt abolished interleukin-4-stimulated or transforming growth factor (TGF)-ß1-stimulated macrophage M2 polarization. Furthermore, deletion of Rictor downregulated TGF-ß1-stimulated upregulation of multiple profibrotic cytokines, including platelet-derived growth factor, vascular endothelial growth factor and connective tissue growth factor, in BMMs. Conditioned medium from TGF-ß1-pretreated Rictor-/- macrophages stimulated fibroblast activation less efficiently than that from TGF-ß1-pretreated Rictor+/+ macrophages. These results demonstrate that Rictor/mTORC2 signalling can promote macrophage activation and kidney fibrosis. Targeting this signalling pathway in macrophages may shine light on ways to protect against kidney fibrosis in patients with chronic kidney diseases. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Proteínas Portadoras/fisiología , Riñón/patología , Activación de Macrófagos/fisiología , Complejos Multiproteicos/fisiología , Serina-Treonina Quinasas TOR/fisiología , Animales , Proteínas Portadoras/metabolismo , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Células Cultivadas , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Fibrosis , Riñón/metabolismo , Macrófagos/metabolismo , Masculino , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones Endogámicos C57BL , Proteína Asociada al mTOR Insensible a la Rapamicina , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
14.
Exp Cell Res ; 345(2): 206-17, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27321958

RESUMEN

Arterial medial calcification (AMC) is prevalent in patients with chronic kidney disease (CKD) and contributes to elevated risk of cardiovascular events and mortality. Vascular smooth muscle cells (VSMCs) to osteogenic transdifferentiation (VOT) in a high-phosphate environment is involved in the pathogenesis of AMC in CKD. WNT/ß-catenin signaling is indicated to play a crucial role in osteogenesis via promoting Runx2 expression in osteoprogenitor cells, however, its role in Runx2 regulation and VOT remains incompletely clarified. In this study, Runx2 was induced and ß-catenin was activated by high-phosphate in VSMCs. Two forms of active ß-catenin, dephosphorylated on Ser37/Thr41 and phosphorylated on Ser675 sites, were upregulated by high-phosphate. Activation of ß-catenin, through ectopic expression of stabilized ß-catenin, inhibition of GSK-3ß, or WNT-3A protein, induced Runx2 expression, whereas blockade of WNT/ß-catenin signaling with Porcupine (PORCN) inhibitor or Dickkopf-1 (DKK1) protein inhibited Runx2 induction by high-phosphate. WNT-3A promoted osteocalcin expression and calcium deposition in VSMCs, whereas DKK1 ameliorated calcification of VSMCs induced by high-phosphate. Two functional T cell factor (TCF)/lymphoid enhancer-binding factor binding sites were identified in the promoter region of Runx2 gene in VSMCs, which interacted with TCF upon ß-catenin activation. Site-directed mutation of each of them attenuated Runx2 response to ß-catenin, and deletion or destruction of both of them completely abolished this responsiveness. In the aortic tunica media of rats with chronic renal failure, followed by AMC, Runx2 and ß-catenin was induced, and the Runx2 mRNA level was positively associated with the abundance of phosphorylated ß-catenin (Ser675). Collectively, our study suggested that high-phosphate may activate WNT/ß-catenin signaling through different pathways, and the activated WNT/ß-catenin signaling, through direct downstream target Runx2, could play an important role in promoting VOT and AMC.


Asunto(s)
Calcificación Fisiológica , Transdiferenciación Celular/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Regulación de la Expresión Génica , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Osteogénesis/genética , Vía de Señalización Wnt , Animales , Aorta/metabolismo , Secuencia de Bases , Sitios de Unión , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/genética , Transdiferenciación Celular/efectos de los fármacos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glicerofosfatos/farmacología , Fallo Renal Crónico/patología , Miocitos del Músculo Liso/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Ratas Sprague-Dawley , Factores de Transcripción TCF/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética , beta Catenina/metabolismo
15.
Am J Physiol Renal Physiol ; 310(1): F27-40, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26469975

RESUMEN

Renal fibrosis is an inevitable outcome of chronic kidney disease (CKD). Erythropoietin (EPO) has been recently reported to be able to mitigate renal fibrosis. The mechanism underlying the protective effect of EPO, however, remains elusive. In the present study, employing a mouse model of renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction (UUO), we demonstrated that EPO markedly reduced the disruption of the tubular basement membrane (TBM) through attenuating the activation of tissue plasminogen activator (tPA) and matrix metalloproteinase 9 (MMP9), the major matrix proteolytic network in the obstructed kidney. Instead of acting directly on tPA in the kidney, EPO strongly increased the level of circulating microRNA (miR)-144, which was delivered to the injured renal fibroblasts via extracellular vesicles (EVs) to target the tPA 3'-untranslated region and suppress tPA expression. The protective effect of EPO on mouse TBM was inhibited by miR-144 antagomir. Furthermore, in vitro results confirmed that EPO could stimulate bone marrow-derived Sca-1(+)CD44(+)CD11b(-)CD19(-) cells to secrete miR-144-containing EVs, which markedly suppressed tPA expression, as well as metalloproteinase 9 (MMP9) level and activity, in cultured renal fibroblasts. In conclusion, our study provides the first evidence that EPO protects mouse renal TBM through promoting bone marrow cells to generate and secrete miR-144, which, in turn, is efficiently delivered into the mouse kidney via EVs to inhibit the activation of the tPA/MMP9-mediated proteolytic network. This finding thus suggests that EPO, a hormone widely used to treat anemia in CKD, is a potential therapeutic strategy for renal fibrosis.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Eritropoyetina/farmacología , Vesículas Extracelulares/efectos de los fármacos , Membrana Basal Glomerular/efectos de los fármacos , Enfermedades Renales/prevención & control , Túbulos Renales/efectos de los fármacos , MicroARNs/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Obstrucción Ureteral/tratamiento farmacológico , Regiones no Traducidas 3' , Animales , Sitios de Unión , Células de la Médula Ósea/enzimología , Línea Celular , Citoprotección , Modelos Animales de Enfermedad , Activación Enzimática , Represión Enzimática , Vesículas Extracelulares/enzimología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibrosis , Membrana Basal Glomerular/enzimología , Membrana Basal Glomerular/patología , Enfermedades Renales/enzimología , Enfermedades Renales/genética , Enfermedades Renales/patología , Túbulos Renales/enzimología , Túbulos Renales/patología , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , MicroARNs/sangre , MicroARNs/genética , Ratas , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Activador de Tejido Plasminógeno/genética , Obstrucción Ureteral/enzimología , Obstrucción Ureteral/genética , Obstrucción Ureteral/patología
16.
J Cell Sci ; 127(Pt 20): 4494-506, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25107369

RESUMEN

Tubular epithelial cell apoptosis contributes to tubulointerstitial fibrosis but its regulation remains unclear. Here, in fibrotic kidney induced by unilateral ureteral obstruction (UUO), we demonstrate that miR-34a is markedly upregulated in tubulointerstitial spaces and microvesicles isolated from obstructed kidney. However, miR-34a is not de novo synthesized by proximal tubular epithelial cells but by fibroblasts after incubation with TGF-ß1. miR-34a is markedly upregulated in microvesicles isolated from the cell culture medium of TGF-ß1-treated fibroblasts. These microvesicles act as a vector for delivery of upregulated miR-34a from fibroblasts to tubular cells. The fibroblast-derived miR-34a-containing microvesicles induce the apoptosis of tubular cells. The exogenous miR-34a regulates tubular apoptosis by modulating the expression of the anti-apoptotic protein Bcl-2. Moreover, injection of exogenous miR-34a-containing microvesicles enhances tubular cell apoptosis in mice. This study suggests that secreted fibroblast miR-34a transported by microvesicles induces tubular cell apoptosis in obstructed kidney. This study reveals a new mechanism whereby microvesicle-mediated communication of miRNA between fibroblasts and tubular cells is involved in regulating tubular cell apoptosis, which might provide new therapeutic targets for renal tubulointerstitial fibrosis.


Asunto(s)
Apoptosis , Túbulos Renales/patología , MicroARNs/metabolismo , Nefritis Intersticial/patología , Vesículas Secretoras/metabolismo , Animales , Comunicación Celular , Línea Celular , Fibroblastos/fisiología , Fibrosis , Regulación de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos , MicroARNs/genética , Terapia Molecular Dirigida , Nefritis Intersticial/genética , Nefritis Intersticial/terapia , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Factor de Crecimiento Transformador beta1/inmunología
17.
Kidney Int ; 88(3): 515-27, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25970154

RESUMEN

The mammalian target of rapamycin (mTOR) was recently identified in two structurally distinct multiprotein complexes: mTORC1 and mTORC2. Previously, we found that Rictor/mTORC2 protects against cisplatin-induced acute kidney injury, but the role and mechanisms for Rictor/mTORC2 in TGFß1-induced fibroblast activation and kidney fibrosis remains unknown. To study this, we initially treated NRK-49F cells with TGFß1 and found that TGFß1 could activate Rictor/mTORC2 signaling in cultured cells. Blocking Rictor/mTORC2 signaling with Rictor or Akt1 small interfering RNAs markedly inhibited TGFß1-induced fibronection and α-smooth muscle actin expression. Ensuing western blotting or immunostaining results showed that Rictor/mTORC2 signaling was activated in kidney interstitial myofibroblasts from mice with unilateral ureteral obstruction. Next, a mouse model with fibroblast-specific deletion of Rictor was generated. These knockout mice were normal at birth and had no obvious kidney dysfunction or kidney morphological abnormality within 2 months of birth. Compared with control littermates, the kidneys of Rictor knockout mice developed less interstitial extracellular matrix deposition and inflammatory cell infiltration at 1 or 2 weeks after ureteral obstruction. Thus our study suggests that Rictor/mTORC2 signaling activation mediates TGFß1-induced fibroblast activation and contributes to the development of kidney fibrosis. This may provide a therapeutic target for chronic kidney diseases.


Asunto(s)
Proteínas Portadoras/metabolismo , Fibroblastos/efectos de los fármacos , Enfermedades Renales/enzimología , Riñón/efectos de los fármacos , Complejos Multiproteicos/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Actinas/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Fibroblastos/enzimología , Fibroblastos/patología , Fibronectinas/metabolismo , Fibrosis , Riñón/enzimología , Riñón/patología , Enfermedades Renales/etiología , Enfermedades Renales/patología , Masculino , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/efectos de los fármacos , Miofibroblastos/enzimología , Miofibroblastos/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Proteína Asociada al mTOR Insensible a la Rapamicina , Factores de Tiempo , Transfección , Obstrucción Ureteral/complicaciones
18.
Cell Physiol Biochem ; 36(5): 1688-98, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26196155

RESUMEN

BACKGROUND: Uncoupling protein 2 (UCP2) is critical in regulating energy metabolism. Due to the significant change in energy metabolism of myocardium upon pressure overload, we hypothesize that UCP2 could contribute to the etiology of cardiac hypertrophy. METHODS: Adult male C57BL/6J mice were subjected to pressure overload by using transverse aortic constriction (TAC), and then received genipin (a UCP2 selective inhibitor; 25 mg/kg/d, ip) or vehicle for three weeks prior to histologic assessment of myocardial hypertrophy. ATP concentration, ROS level, and myocardial apoptosis were also examined. A parallel set of experiments was also conducted in UCP2-/- mice. RESULTS: TAC induced left ventricular hypertrophy, as reflected by increased ventricular weight/thickness and increased size of myocardial cell (vs. sham controls). ATP concentration was decreased; ROS level was increased. Apoptosis and fibrosis markers were increased. TAC increased mitochondrial UCP2 expression in the myocardium at both mRNA and protein levels. Genipin treatment attenuated cardiac hypertrophy and the histologic/biochemical changes described above. Hypertrophy and associated changes induced by TAC in UCP2-/- mice were much less pronounced than in WT mice. CONCLUSIONS: Blocking UCP2 expression attenuates cardiac hypertrophy induced by pressure overload.


Asunto(s)
Aorta/patología , Cardiomegalia/prevención & control , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Cardiomegalia/etiología , Constricción Patológica , Canales Iónicos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/genética , Especies Reactivas de Oxígeno/metabolismo , Proteína Desacopladora 2
19.
Exp Cell Res ; 322(2): 290-301, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24424244

RESUMEN

Podocyte apoptosis is a major factor inducing podocyte depletion that predicts the progressive course of glomerulosclerosis. However, the molecular mechanisms underlying podocyte apoptosis are still not well understood. Autophagy is a lysosomal degradation system involving the degradation and recycling of obsolete, damaged, or harmful cytoplasmic materials and organelles. Recent advances in the understanding of the molecular processes contributing to autophagy have provided insight into the relationship between autophagy and apoptosis. However, their cross-talk remains largely obscure until now. Here, we found that podocytes both in vivo and in vitro always exhibited high basal levels of autophagy, whereas autophagy inhibition could induce podocyte apoptosis, suggesting the pro-survival role of autophagy in podocytes. Besides, we found that autophagy inhibition by 3-methyladenine (3-MA) could induce the activation of endoplasmic reticulum stress even without any external stimulations, whereas knockdown of CHOP could effectively improve podocyte apoptosis and down-regulated expression of slit-diaphragm proteins induced by autophagy inhibition. Collectively, this study demonstrated that autophagy might act as a crucial regulatory mechanism of apoptotic cell death by modulating the balance between the pro-survival pathway and the pro-apoptotic pathway of endoplasmic reticulum stress, which might provide a novel mechanistic insight into the interface between autophagy and apoptosis in the progression of podocyte injury.


Asunto(s)
Adenina/análogos & derivados , Apoptosis/efectos de los fármacos , Autofagia , Nefropatías Diabéticas/patología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Podocitos/patología , Adenina/farmacología , Adulto , Animales , Western Blotting , Estudios de Casos y Controles , Células Cultivadas , Nefropatías Diabéticas/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Masculino , Ratones , Microscopía Fluorescente , Persona de Mediana Edad , Podocitos/efectos de los fármacos , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción CHOP/antagonistas & inhibidores , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo
20.
Exp Cell Res ; 322(2): 302-12, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24486760

RESUMEN

OBJECTIVES: Vascular calcification is highly prevalent in patients with chronic kidney disease (CKD) and contributes to increased risk of cardiovascular disease and mortality. Accumulated evidences suggested that vascular smooth muscle cells (VSMCs) to osteoblast-like cells transdifferentiation (VOT) plays a crucial role in promoting vascular calcification. MicroRNAs (miRNAs) are a novel class of small RNAs that negatively regulate gene expression via repression of the target mRNAs. In the present work, we sought to determine the role of miRNAs in VSMCs phenotypic transition and calcification induced by ß-glycerophosphoric acid. APPROACH AND RESULTS: Primary cultured rat aortic VSMCs were treated with ß-glycerophosphoric acid for different periods of time. In VSMCs, after ß-glycerophosphoric acid treatment, the expressions of cbf ß1, osteocalcin and osteopontin were significantly increased and SM-22ß expression was decreased. ALP activity was induced by ß-glycerophosphoric acid in a time or dose dependent manner. Calcium deposition was detected in VSMCs incubated with calcification media; then, miR-125b expression was detected by real-time RT PCR. miR-125b expression was significantly decreased in VSMCs after incubated with ß-glycerophosphoric acid. Overexpression of miR-125b could inhibit ß-glycerophosphoric acid-induced osteogenic markers expression and calcification of VSMCs whereas knockdown of miR-125b promoted the phenotypic transition of VSMCs and calcification. Moreover, miR-125b targeted Ets1 and regulated its protein expression in VSMCs. Downregulating Ets1 expression by its siRNA inhibited ß-glycerophosphoric acid-induced the VSMCs phenotypic transition and calcification. CONCLUSION: Our study suggests that down-regulation of miR-125b after ß-glycerophosphoric acid treatment facilitates VSMCs transdifferentiation and calcification through targeting Ets1.


Asunto(s)
Calcificación Fisiológica , Transdiferenciación Celular/efectos de los fármacos , Glicerofosfatos/farmacología , MicroARNs/genética , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Aorta/citología , Aorta/efectos de los fármacos , Aorta/metabolismo , Western Blotting , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Microscopía Fluorescente , Músculo Liso Vascular/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Proteína Proto-Oncogénica c-ets-1/genética , ARN Mensajero/genética , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA