Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Kidney Int ; 103(5): 886-902, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36804379

RESUMEN

Progressive fibrosis is a hallmark of chronic kidney disease, but we lack effective treatments to halt this destructive process. Micropeptides (peptides of no more than 100 amino acids) encoded by small open reading frames represent a new class of eukaryotic regulators. Here, we describe that the micropeptide regulator of ß-oxidation (MOXI) regulates kidney fibrosis. MOXI expression was found to be up-regulated in human fibrotic kidney disease, and this correlated with the degree of fibrosis and loss of kidney function. MOXI was expressed in the cytoplasm and mitochondria of cultured tubular epithelial cells and translocated to the nucleus upon Transforming Growth Factor-ß1 stimulation. Deletion of Moxi protected mice against fibrosis and inflammation in the folic acid and unilateral ureteral obstruction models. As a potential molecular therapy, treatment with an antisense MOXI oligonucleotide effectively knocked-down MOXI expression and protected against kidney fibrosis in both models. Bimolecular fluorescence complementation identified the enzyme N-acetyltransferase 14 (Nat14) and transcription factor c-Jun as MOXI binding partners. The MOXI/Nat14/c-Jun complex enhances basal and Transforming Growth Factor-ß1 induced collagen I gene promoter activity. Phosphorylation at T49 is required for MOXI nuclear localization and for complex formation with Nat14 and c-Jun. Furthermore, mice with a MoxiT49A point mutation were protected in the models of kidney fibrosis. Thus, our studies demonstrate a key role for the micropeptide MOXI in kidney fibrosis and identify a new function of MOXI in forming a transcriptional complex with Nat14 and c-Jun.


Asunto(s)
Enfermedades Renales , Obstrucción Ureteral , Animales , Humanos , Ratones , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Fibrosis , Riñón/patología , Enfermedades Renales/patología , Mitocondrias/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/genética , Obstrucción Ureteral/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Micropéptidos
2.
Development ; 147(10)2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32366677

RESUMEN

Thanks to many advances in genetic manipulation, mouse models have become very powerful in their ability to interrogate biological processes. In order to precisely target expression of a gene of interest to particular cell types, intersectional genetic approaches using two promoter/enhancers unique to a cell type are ideal. Within these methodologies, variants that add temporal control of gene expression are the most powerful. We describe the development, validation and application of an intersectional approach that involves three transgenes, requiring the intersection of two promoter/enhancers to target gene expression to precise cell types. Furthermore, the approach uses available lines expressing tTA/rTA to control the timing of gene expression based on whether doxycycline is absent or present, respectively. We also show that the approach can be extended to other animal models, using chicken embryos. We generated three mouse lines targeted at the Tigre (Igs7) locus with TRE-loxP-tdTomato-loxP upstream of three genes (p21, DTA and Ctgf), and combined them with Cre and tTA/rtTA lines that target expression to the cerebellum and limbs. Our tools will facilitate unraveling biological questions in multiple fields and organisms.


Asunto(s)
Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen , Transgenes , Animales , Cerebelo , Embrión de Pollo , Doxiciclina/farmacología , Extremidades , Femenino , Masculino , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Transactivadores/genética , Transcripción Genética
3.
EMBO Rep ; 21(2): e48781, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31916354

RESUMEN

Diabetic nephropathy (DN) is the leading cause of end-stage kidney disease. TGF-ß1/Smad3 signalling plays a major pathological role in DN; however, the contribution of Smad4 has not been examined. Smad4 depletion in the kidney using anti-Smad4 locked nucleic acid halted progressive podocyte damage and glomerulosclerosis in mouse type 2 DN, suggesting a pathogenic role of Smad4 in podocytes. Smad4 is upregulated in human and mouse podocytes during DN. Conditional Smad4 deletion in podocytes protects mice from type 2 DN, independent of obesity. Mechanistically, hyperglycaemia induces Smad4 localization to mitochondria in podocytes, resulting in reduced glycolysis and oxidative phosphorylation and increased production of reactive oxygen species. This operates, in part, via direct binding of Smad4 to the glycolytic enzyme PKM2 and reducing the active tetrameric form of PKM2. In addition, Smad4 interacts with ATPIF1, causing a reduction in ATPIF1 degradation. In conclusion, we have discovered a mitochondrial mechanism by which Smad4 causes diabetic podocyte injury.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Podocitos , Animales , Diabetes Mellitus/metabolismo , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Glucólisis/genética , Riñón , Ratones , Podocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo
4.
Differentiation ; 92(3): 102-107, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27262400

RESUMEN

Renal fibrosis is a common characteristic of chronic kidney disease (CKD). Aberrant and excessive depositions of extracellular matrix (ECM) proteins in both glomeruli and interstitial regions are typical hallmarks of renal fibrosis and amplify the severity of kidney injury. To date, an approved therapy specifically targeted to renal fibrosis is needed to mitigate or even retard renal fibrosis. Recent findings have identified a unique population of myofibroblasts as a primary source of ECM in scar tissue formation. However, the origin of myofibroblasts in renal fibrosis remains the subject of controversial debates. The advancement in lineage tracing and immunofluorescent microscopy technologies have suggested that myofibroblasts may arise from a number of sources such as activated renal fibroblasts, pericytes, epithelial-to-mesenchymal transition (EMT), endothelial-to-mesenchymal transition (EndoMT), bone marrow derived cells and fibrocytes. Recent studies also indicate that multiple ligands of TGF-ß/Smads are the direct mediators for renal fibrosis. Consistently, inhibition of the TGF-ß/Smads signaling pathway using various strategies significantly reduce renal fibrotic lesions and ameliorate kidney injury, suggesting that targeting the TGF-ß/Smads signaling pathway could be a new strategy for effective therapies. In this review, we will briefly discuss the diverse origins of myofibroblasts and molecular pathways triggering renal fibrosis. Prospective therapeutic approaches based on those molecular mechanisms will hopefully offer exciting insights in the development of new therapeutic interventions for patients in the near future.


Asunto(s)
Fibroblastos/metabolismo , Fibrosis/patología , Enfermedades Renales/patología , Riñón/metabolismo , Miofibroblastos/patología , Animales , Transición Epitelial-Mesenquimal/fisiología , Fibrosis/diagnóstico , Humanos , Riñón/patología , Enfermedades Renales/metabolismo
5.
Kidney Int ; 88(6): 1323-1335, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26221756

RESUMEN

Transforming growth factor-ß1 (TGF-ß1)/Smad signaling has a central role in the pathogenesis of renal fibrosis. Smad3 and Smad4 are pro-fibrotic, while Smad2 is anti-fibrotic. However, these Smads form heterogeneous complexes, the functions of which are poorly understood. Here we studied Smad complex function in renal fibrosis using the mouse model of unilateral ureteric obstruction. Mice heterozygous for Smad3/4 (Smad3/4+/-) exhibited substantial protection from renal fibrosis through day 7 of obstruction, whereas Smad2/3+/- and Smad2/4+/- mice showed only modest protection. Formation of Smad3/Smad4/CDK9 complexes was an early event following obstruction in wild-type mice, which involved nuclear phosphorylation of the linker regions of Smad3. Significantly, Smad3 or Smad4 deficiency decreased the formation of Smad4/CDK9 or Smad3/CDK9 complex, Smad3 linker phosphorylation, and fibrosis but at different degrees. In vitro, TGF-ß1 stimulation of collagen I promoter activity involved formation of Smad3/Smad4/CDK9 complexes, and overexpression of each component gave additive increases in collagen promoter activity. Co-administration of a CDK9 inhibitor and Smad3-specific inhibition achieved better protection from TGF-ß1-induced fibrotic response in vitro and renal interstitial fibrosis in vivo. Thus formation of Smad3/Smad4/CDK9 complex drives renal fibrosis during ureteral obstruction. Formation of this complex represents a novel target for antifibrotic therapies.

6.
Kidney Int ; 88(2): 286-98, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25945408

RESUMEN

Signaling by TGF-ß/Smad3 plays a key role in renal fibrosis. As obesity is one of the major risk factors of chronic and end-stage renal disease, we studied the role of Smad3 signaling in the pathogenesis of obesity-related renal disease. After switching to a high fat diet, the onset of Smad3 C-terminal phosphorylation, increase in albuminuria, and the early stages of peripheral and renal insulin resistance occurred at 1 day, and 4 and 8 weeks, respectively, in C57BL/6 mice. The loss of synaptopodin, a functional marker of podocytes, and phosphorylation of the Smad3 linker region (T179 and S213) appeared after 4 weeks of the high fat diet. This suggests a temporal pattern of Smad3 signaling activation leading to kidney injury and subsequent insulin resistance in the development of obesity-related renal disease. In vivo, Smad3 knockout attenuated the high fat diet-induced proteinuria, renal fibrosis, overall podocyte injury, and mitochondrial dysfunction in podocytes. In vitro palmitate caused a rapid activation of Smad3 in 30 min, loss of synaptopodin in 2 days, and impaired insulin signaling in 3 days in isolated mouse podocytes. Blockade of either Smad3 phosphorylation by SIS3 (a Smad3 inhibitor) or T179 phosphorylation by flavopiridol (a CDK9 inhibitor) prevented the palmitate-induced loss of synaptopodin and mitochondrial function in podocytes. Thus, Smad3 signaling plays essential roles in obesity-related renal disease and may be a novel therapeutic target.


Asunto(s)
Obesidad/complicaciones , Obesidad/metabolismo , Podocitos/metabolismo , Transducción de Señal , Proteína smad3/deficiencia , Animales , Células Cultivadas , Grasas de la Dieta/administración & dosificación , Inhibidores Enzimáticos/farmacología , Fibrosis , Flavonoides/farmacología , Técnicas de Silenciamiento del Gen , Insulina/metabolismo , Resistencia a la Insulina , Isoquinolinas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ácido Palmítico/farmacología , Fosforilación/efectos de los fármacos , Piperidinas/farmacología , Podocitos/efectos de los fármacos , Factores Protectores , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Pirroles/farmacología , Transducción de Señal/efectos de los fármacos , Proteína smad3/genética
7.
Am J Pathol ; 184(4): 944-952, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24485922

RESUMEN

Transforming growth factor-ß (TGF-ß) promotes tissue fibrosis via receptor-mediated phosphorylation of the receptor-activated Smad2/3, together with Smad4. Of these, Smad3 plays a major profibrotic role in mouse models of tissue fibrosis. Transcriptional activity of the Smad3 protein is regulated by phosphorylation of residues in the C-terminal domain and the linker region. Herein, we examined the role of a novel phosphorylation site within the MH2 domain (T388) in the regulation of Smad3 activity. Confocal microscopy using an Smad3 phosphorylated T388-specific antibody identified phosphorylation of Smad3 T388 in myofibroblasts and tubular epithelial cells in human focal and segmental glomerulosclerosis and mouse models of unilateral ureteric obstruction and diabetic nephropathy, whereas phosphorylated T388 was largely absent in normal kidney. In vitro, TGF-ß1 induced phosphorylation of Smad3 T388 in a biphasic pattern. A point mutation of T388/V in an Smad3 construct demonstrated that phosphorylation of T388 promotes Smad3 binding to Smad4 and CDK8, but was not necessary for nuclear translocation. Furthermore, T388 phosphorylation was required for TGF-ß-induced collagen I gene promoter activity and extracellular matrix production in cultured fibroblasts. In conclusion, our study identifies phosphorylation of T388 in the Smad3 MH2 domain as an important mechanism that regulates the profibrotic TGF-ß/Smad3 signaling pathway, which has direct relevance to human and experimental fibrotic kidney disease.


Asunto(s)
Enfermedades Renales/metabolismo , Transducción de Señal/fisiología , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Western Blotting , Fibrosis/metabolismo , Humanos , Inmunoprecipitación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Fosforilación , Estructura Terciaria de Proteína , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Nat Commun ; 15(1): 2940, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38580631

RESUMEN

A major question in developmental and regenerative biology is how organ size and architecture are controlled by progenitor cells. While limb bones exhibit catch-up growth (recovery of a normal growth trajectory after transient developmental perturbation), it is unclear how this emerges from the behaviour of chondroprogenitors, the cells sustaining the cartilage anlagen that are progressively replaced by bone. Here we show that transient sparse cell death in the mouse fetal cartilage is repaired postnatally, via a two-step process. During injury, progression of chondroprogenitors towards more differentiated states is delayed, leading to altered cartilage cytoarchitecture and impaired bone growth. Then, once cell death is over, chondroprogenitor differentiation is accelerated and cartilage structure recovered, including partial rescue of bone growth. At the molecular level, ectopic activation of mTORC1 correlates with, and is necessary for, part of the recovery, revealing a specific candidate to be explored during normal growth and in future therapies.


Asunto(s)
Cartílago , Condrocitos , Animales , Ratones , Condrocitos/metabolismo , Diferenciación Celular , Huesos , Muerte Celular
9.
J Clin Invest ; 134(10)2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38625739

RESUMEN

Renal interstitial fibrosis is an important mechanism in the progression of chronic kidney disease (CKD) to end-stage kidney disease. However, we lack specific treatments to slow or halt renal fibrosis. Ribosome profiling identified upregulation of a secreted micropeptide, C4orf48 (Cf48), in mouse diabetic nephropathy. Cf48 RNA and protein levels were upregulated in tubular epithelial cells in human and experimental CKD. Serum Cf48 levels were increased in human CKD and correlated with loss of kidney function, increasing CKD stage, and the degree of active interstitial fibrosis. Cf48 overexpression in mice accelerated renal fibrosis, while Cf48 gene deletion or knockdown by antisense oligonucleotides significantly reduced renal fibrosis in CKD models. In vitro, recombinant Cf48 (rCf48) enhanced TGF-ß1-induced fibrotic responses in renal fibroblasts and epithelial cells independently of Smad3 phosphorylation. Cellular uptake of Cf48 and its profibrotic response in fibroblasts operated via the transferrin receptor. RNA immunoprecipitation-sequencing identified Cf48 binding to mRNA of genes involved in the fibrotic response, including Serpine1, Acta2, Ccn2, and Col4a1. rCf48 binds to the 3'UTR of Serpine1 and increases mRNA half-life. We identify the secreted Cf48 micropeptide as a potential enhancer of renal fibrosis that operates as an RNA-binding peptide to promote the production of extracellular matrix.


Asunto(s)
Nefropatías Diabéticas , Fibrosis , Proteínas del Tejido Nervioso , Insuficiencia Renal Crónica , Animales , Humanos , Masculino , Ratones , Regiones no Traducidas 3' , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/genética , Riñón/metabolismo , Riñón/patología , Ratones Noqueados , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteína smad3/metabolismo , Proteína smad3/genética , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo
10.
J Pathol ; 228(4): 506-19, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22610993

RESUMEN

Resolvin E1 (RvE1) is a naturally occurring lipid-derived mediator generated during the resolution of inflammation. The anti-inflammatory effects of RvE1 have been demonstrated in a variety of disease settings; however, it is not known whether RvE1 may also exert direct anti-fibrotic effects. We examined the potential anti-fibrotic actions of RvE1 in the mouse obstructed kidney-a model in which tissue fibrosis is driven by unilateral ureteric obstruction (UUO), an irreversible, non-immune insult. Administration of RvE1 (300 ng/day) to mice significantly reduced accumulation of α-smooth muscle actin (SMA)(+) myofibroblasts and the deposition of collagen IV on day 6 after UUO. This protective effect was associated with a marked reduction of myofibroblast proliferation on days 2, 4 and 6 after UUO. RvE1 treatment also inhibited production of the major fibroblast mitogen, platelet-derived growth factor-BB (PDGF-BB), in the obstructed kidney. Acute resolvin treatment over days 2-4 after UUO also had a profound inhibitory effect upon myofibroblast proliferation without affecting the PDGF expression, suggesting a direct effect upon fibroblast proliferation. In vitro studies established that RvE1 can directly inhibit PDGF-BB-induced proliferation in primary mouse fibroblasts. RvE1 induced transient, but not sustained, activation of the pro-proliferative ERK and AKT signalling pathways. Of note, RvE1 inhibited the sustained activation of ERK and AKT pathways seen in response to PDGF stimulation, thereby preventing up-regulation of molecules required for progression through the cell cycle (c-Myc, cyclin D) and down-regulation of inhibitors of cell cycle progression (p21, cip1). Finally, siRNA-based knock-down studies showed that the RvE1 receptor, ChemR23, is required for the anti-proliferative actions of RvE1 in cultured fibroblasts. In conclusion, this study demonstrates that RvE1 can inhibit fibroblast proliferation in vivo and in vitro, identifying RvE1 as a novel anti-fibrotic therapy.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/análogos & derivados , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Obstrucción Ureteral/tratamiento farmacológico , Obstrucción Ureteral/patología , Animales , Becaplermina , Proliferación Celular/efectos de los fármacos , Ácido Eicosapentaenoico/farmacología , Fibrosis/tratamiento farmacológico , Fibrosis/patología , Riñón/efectos de los fármacos , Riñón/patología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-sis/farmacología
11.
Am J Pathol ; 177(3): 1065-71, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20651248

RESUMEN

Transforming growth factor-beta1 (TGF-beta1) promotes tissue fibrosis through the Smad3 signaling pathway. While phosphorylation is known to regulate Smad3 function, recent in vitro studies have suggested that acetylation may also regulate Smad3 function. This study investigated Smad3 acetylation in renal fibrosis. TGF-beta1 stimulation of renal fibroblasts and tubular epithelial cells induced Smad3 acetylation and phosphorylation. Resveratrol, an activator of the Nicotinamide adenine dinucleotide (NAD) dependent protein deacetylase SIRT1, reversed acetylation but not phosphorylation of Smad3 and inhibited TGF-beta1-induced up-regulation of collagen IV and fibronectin mRNA levels. Knockdown of SIRT1 expression abolished the inhibitory effect of resveratrol, and co-immunoprecipitation studies provide direct evidence of an interaction between acetylated Smad3 and SIRT1. The role of Smad3 acetylation in renal fibrosis was then examined in the unilateral ureteric obstruction (UUO) model. Immunoprecipitation studies showed acetylation and phosphorylation of Smad3 by day 2 UUO, which was sustained to day 7 in association with development of interstitial fibrosis. Resveratrol inhibited acetylation but not phosphorylation of Smad3 at day 2 UUO, and resveratrol treatment inhibited interstitial fibrosis at day 7 UUO. In conclusion, these studies support a pathological role for Smad3 acetylation in renal fibrosis and suggest that deacetylation of Smad3 may be a novel therapeutic target for fibrotic disease.


Asunto(s)
Fibroblastos/efectos de los fármacos , Riñón/efectos de los fármacos , Proteína smad3/metabolismo , Estilbenos/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Acetilación/efectos de los fármacos , Análisis de Varianza , Animales , Western Blotting , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis/metabolismo , Fibrosis/patología , Inmunoprecipitación , Riñón/metabolismo , Riñón/patología , Ratones , Microscopía Confocal , Resveratrol , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Sirtuina 1/genética , Sirtuina 1/metabolismo , Proteína smad3/genética , Factor de Crecimiento Transformador beta1/genética
12.
Front Cell Dev Biol ; 9: 736574, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34513850

RESUMEN

The characterization of developmental phenotypes often relies on the accurate linear measurement of structures that are small and require laborious preparation. This is tedious and prone to errors, especially when repeated for the multiple replicates that are required for statistical analysis, or when multiple distinct structures have to be analyzed. To address this issue, we have developed a pipeline for characterization of long-bone length using X-ray microtomography (XMT) scans. The pipeline involves semi-automated algorithms for automatic thresholding and fast interactive isolation and 3D-model generation of the main limb bones, using either the open-source ImageJ plugin BoneJ or the commercial Mimics Innovation Suite package. The tests showed the appropriate combination of scanning conditions and analysis parameters yields fast and comparable length results, highly correlated with the measurements obtained via ex vivo skeletal preparations. Moreover, since XMT is not destructive, the samples can be used afterward for histology or other applications. Our new pipelines will help developmental biologists and evolutionary researchers to achieve fast, reproducible and non-destructive length measurement of bone samples from multiple animal species.

13.
Am J Pathol ; 175(4): 1380-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19729486

RESUMEN

Diabetic nephropathy is the leading cause of chronic renal failure. Myofibroblasts play a major role in the synthesis and secretion of extracellular matrix in diabetic renal fibrosis. Increasing evidence suggests that endothelial cells may undergo endothelial-myofibroblast transition under physiological and pathophysiological circumstances. Therefore, this study investigates whether endothelial-myofibroblast transition occurs and contributes to the development of diabetic renal interstitial fibrosis. Diabetes was induced by administration of streptozotocin to Tie2-Cre;LoxP-EGFP mice, an endothelial lineage-traceable mouse line generated by crossbreeding B6.Cg-Tg(Tek-cre)12F1v/J mice with B6.Cg-Tg(ACTB-Bgeo/GFP)21Lbe/J mice. The endothelial-myofibroblast transition was also studied in MMECs (a mouse pancreatic microvascular endothelial cell line) and primary cultures of CD31+/EYFP- (enhanced yellow fluorescent protein) endothelial cells isolated from adult normal alpha-smooth muscle actin promoter-driven-EYFP (alpha-SMA/EYFP) mouse kidneys. Confocal microscopy demonstrated that 10.4 +/- 4.2 and 23.5 +/- 7.4% of renal interstitial myofibroblasts (alpha-SMA+) in 1- and 6-month streptozotocin-induced diabetic kidneys were of endothelial origin (EGFP+/alpha-SMA+ cells), compared with just 0.2 +/- 0.1% of myofibroblasts in vehicle-treated Tie2-Cre;LoxP-EGFP mice (P < 0.01). Confocal microscopy and real-time PCR showed that transforming growth factor (TGF)-beta1 induced de novo expression of alpha-SMA and loss of expression of VE-cadherin and CD31 in MMECs and primary cultures of renal endothelial cells in a time- and dose-dependent fashion. These findings demonstrate that the endothelial-myofibroblast transition occurs and contributes to the early development and progression of diabetic renal interstitial fibrosis and suggest that the endothelial-myofibroblast transition may be a therapeutic target.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/complicaciones , Nefropatías Diabéticas/patología , Endotelio/patología , Fibroblastos/patología , Actinas/metabolismo , Animales , Linaje de la Célula/efectos de los fármacos , Endotelio/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibrosis , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Integrasas/metabolismo , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Ratones , Microscopía Confocal , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor TIE-2 , Estreptozocina , Factor de Crecimiento Transformador beta1/farmacología
14.
Front Pharmacol ; 10: 880, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31447676

RESUMEN

Acute kidney injury leading to chronic kidney disease through tubulointerstitial fibrosis is a major challenge in nephropathy. Several signaling pathways promote interstitial fibrosis; however, effective suppression of fibrosis may require blockade of more than one pathway. This study investigated whether blockade of Smad3 and c-Jun N-terminal kinase (JNK) signaling gives added suppression of interstitial fibrosis in folic acid nephropathy. A single high dose of folic acid (FA) causes acute tubular damage in C57BL/6J mice followed by interstitial fibrosis and chronic renal impairment. Co-activations of Smad3 and JNK signaling occur in both tubular epithelial cells and myofibroblasts in areas of tubulointerstitial damage and fibrosis in both murine FA-induced nephropathy and human IgA nephropathy. Groups of mice were treated with a Smad3 inhibitor (SIS3), a JNK inhibitor (SP600125), or a combination from day 6 after FA administration until being killed on day 28. Each drug efficiently inhibited its specific target (Smad3 phosphorylation or c-Jun phosphorylation) without affecting the other pathway. Given alone, each drug partially reduced renal fibrosis, whereas the combination therapy gave an additive and profound protection from renal fibrosis and improved renal function. Inhibition of Smad3 and/or JNK signaling activities prevented down-regulation of PGC-1α in tubular epithelial cells and up-regulation of PGC-1α in myofibroblasts during FA-induced renal fibrosis and inflammation. The expression of PGC-1α was upregulated in Smad3 -/- NRK52E cells while downregulated in Smad3 -/- NRK49F cells, suggesting that Smad3 signaling may regulate expression of PGC-1α in renal tubular epithelial cells and fibroblasts in distinct fashion. In vivo and cell culture studies also indicate that Smad3 and JNK signaling cooperate to cause mitochondrial dysfunction and cell damage in tubular epithelial cells via direct actions on the transcription of PGC-1α. These pathways also act cooperatively to promote renal fibroblast proliferation in tempo-spatial fashion. In conclusion, we have identified a potential combination therapy for progressive renal fibrosis which operates, in part, through modifying mitochondrial function.

15.
PLoS One ; 8(1): e55027, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23359116

RESUMEN

The role of podocytes in the development and progression of glomerular disease has been extensively investigated in the past decade. However, the importance of glomerular endothelial cells in the pathogenesis of proteinuria and glomerulosclerosis has been largely ignored. Recent studies have demonstrated that endothelial nitric oxide synthatase (eNOS) deficiency exacerbates renal injury in anti-GBM and remnant kidney models and accelerates diabetic kidney damage. Increasing evidence also demonstrates the importance of the glomerular endothelium in preventing proteinuria. We hypothesize that endothelial dysfunction can initiate and promote the development and progression of glomerulopathy. Administration of adriamycin (ADR) to C57BL/6 mice, normally an ADR resistant strain, with an eNOS deficiency induced overt proteinuria, severe glomerulosclerosis, interstitial fibrosis and inflammation. We also examined glomerular endothelial cell and podocyte injury in ADR-induced nephropathy in Balb/c mice, an ADR susceptible strain, by immunostaining, TUNEL and Western blotting. Interestingly, down-regulation of eNOS and the appearance of apoptotic glomerular endothelial cells occurred as early as 24 hours after ADR injection, whilst synaptopodin, a functional podocyte marker, was reduced 7 days after ADR injection and coincided with a significant increase in the number of apoptotic podocytes. Furthermore, conditioned media from mouse microvascular endothelial cells over-expressing GFP-eNOS protected podocytes from TNF-α-induced loss of synaptopodin. In conclusion, our study demonstrated that endothelial dysfunction and damage precedes podocyte injury in ADR-induced nephropathy. Glomerular endothelial cells may protect podocytes from inflammatory insult. Understanding the role of glomerular endothelial dysfunction in the development of kidney disease will facilitate in the design of novel strategies to treat kidney disease.


Asunto(s)
Doxorrubicina/toxicidad , Endotelio Vascular/patología , Glomérulos Renales/patología , Riñón/efectos de los fármacos , Podocitos/patología , Animales , Western Blotting , Medios de Cultivo Condicionados , Endotelio Vascular/enzimología , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Confocal , Óxido Nítrico Sintasa de Tipo III/metabolismo
16.
PLoS One ; 8(12): e84063, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24391884

RESUMEN

Endothelial dysfunction and enhanced transforming growth factor-ß (TGF-ß)/Smad3 signalling are common features of progressive renal fibrosis. This study investigated a potential link between these mechanisms. In unilateral ureteric obstruction (UUO) we observed an acute (6 hr) down-regulation of nitric oxide synthase 3 (NOS3/eNOS) levels and increased phosphorylation of the linker region of Smad3 at T179 and S208 in Smad3/JNK complexes. These events preceded Smad3 C-terminal domain phosphorylation and the induction of myofibroblast proliferation at 48 hrs. Mice deficient in NOS3 showed enhanced myofibroblast proliferation and collagen accumulation compared to wild type mice in a 7 day UUO model. This was associated with enhanced phosphorylation of Smad3 T179 and S208 by 92% and 88%, respectively, whereas Smad3-C-terminal phosphorylation was not affected. Resolvin D1 (RvD1) can suppress renal fibrosis in the UUO model, and further analysis herein showed that RvD1 protected against endothelial dysfunction and suppressed Smad3/JNK complex formation with a consequent reduction in phosphorylation of Smad3 T179 and S208 by 78% and 65%, respectively, while Smad3 C-terminal phosphorylation was unaltered. In vitro, conditioned media from mouse microvascular endothelial cells (MMEC) treated with a general inhibitor of nitric oxide synthase (L-NAME) augmented the proliferation and collagen production of renal fibroblasts (NRK49F cells) compared to control MMEC media and this was associated with increased phosphorylation of JNK and Smad3 T179 and S208, whereas Smad3-C-terminal domain phosphorylation was unaffected. The addition of RvD1 to L-NAME treated MMEC abrogated these effects of the conditioned media on renal fibroblasts. Finally, Smad3 T179/V and S208/A mutations significantly inhibit TGF-ß1 induced up-regulation collagen I promoter. In conclusion, these data suggest that endothelial dysfunction can exacerbate renal interstitial fibrosis through increased fibroblast proliferation and collagen production via enhanced Smad3 linker phosphorylation.


Asunto(s)
Endotelio Vascular/patología , Fibroblastos/patología , Fibrosis/patología , Enfermedades Renales/patología , Óxido Nítrico Sintasa de Tipo III/fisiología , Proteína smad3/metabolismo , Obstrucción Ureteral/patología , Animales , Western Blotting , Proliferación Celular , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Medios de Cultivo Condicionados/farmacología , Ácidos Docosahexaenoicos/farmacología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Inmunoprecipitación , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , Luciferasas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Fosforilación , Regiones Promotoras Genéticas , Ratas , Transducción de Señal , Proteína smad3/genética , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/tratamiento farmacológico , Obstrucción Ureteral/metabolismo
17.
PLoS One ; 8(6): e67471, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23840712

RESUMEN

Resolvin D1 (RvD1) is a lipid-derived mediator generated during the resolution inflammation. While the immunoresolvent effects of Resolvins have been extensively studied in leukocytes, actions of Resolvins on intrinsic kidney cells have received little attention. The podocyte plays a central role in glomerular function, and podocyte damage can lead to proteinuria and glomerulosclerosis. This study examined whether RvD1 has renoprotective effects upon podocytes. We investigated a mouse model of adriamycin (ADR) nephropathy featuring rapid induction of podocyte damage and proteinuria followed by glomerulosclerosis. We identified a progressive loss of synaptopodin expression over a 28 day time-course of ADR nephropathy which was associated with increased acetylation of 14-3-3ß and reduced synaptopodin phosphorylation. Groups of mice were given once daily RvD1 treatment (4 ng/g body weight/day) starting either 30 min (early treatment) or 14 days (late treatment) after ADR injection and continued until mice were killed on day 28. Early, but not late, RvD1 treatment attenuated ADR-induced proteinuria, glomerulosclerosis and tubulointerstitial fibrosis, modified macrophages from an M1 to M2 phenotype. Early RvD1 treatment prevented the down-regulation of synaptopodin expression and changes in 14-3-3ß acetylation and synaptopodin phosphorylation. In a podocyte cell line, RvD1 was shown to prevent rapid TNF-α-induced down-regulation of synaptopodin expression. In transfection studies, TNF-α-induced a decrease in synaptopodin phosphorylation and an increase in acetylation of 14-3-3ß, resulting in disassociation between 14-3-3ß and synaptopodin. RvD1 prevented TNF-α induced post-translational modification of synaptopodin and 14-3-3ß proteins, and maintained the synaptopodin/14-3-3ß interaction. Furthermore, replacement of lysine K51, or K117+K122 in 14-3-3ß with glutamine, to mimic lysine acetylation, significantly reduced the interaction between 14-3-3ß and synaptopodin. In conclusion, our studies provide the first evidence that RvD1 can protect against podocyte damage by preventing down-regulation of synaptopodin through inhibition of 14-3-3ß/synaptopodin dissociation. RvD1 treatment may have potential application in the treatment of chronic kidney disease.


Asunto(s)
Proteínas 14-3-3/genética , Acetilación/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Doxorrubicina/farmacología , Enfermedades Renales/tratamiento farmacológico , Glomérulos Renales/efectos de los fármacos , Podocitos/efectos de los fármacos , Proteínas 14-3-3/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Células HEK293 , Humanos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Glomérulos Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Fosforilación/genética , Podocitos/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/genética , Factor de Necrosis Tumoral alfa/metabolismo
18.
Diabetes ; 59(10): 2612-24, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20682692

RESUMEN

OBJECTIVE: A multicenter, controlled trial showed that early blockade of the renin-angiotensin system in patients with type 1 diabetes and normoalbuminuria did not retard the progression of nephropathy, suggesting that other mechanism(s) are involved in the pathogenesis of early diabetic nephropathy (diabetic nephropathy). We have previously demonstrated that endothelial-mesenchymal-transition (EndoMT) contributes to the early development of renal interstitial fibrosis independently of microalbuminuria in mice with streptozotocin (STZ)-induced diabetes. In the present study, we hypothesized that blocking EndoMT reduces the early development of diabetic nephropathy. RESEARCH DESIGN AND METHODS: EndoMT was induced in a mouse pancreatic microvascular endothelial cell line (MMEC) in the presence of advanced glycation end products (AGEs) and in the endothelial lineage-traceble mouse line Tie2-Cre;Loxp-EGFP by administration of AGEs, with nonglycated mouse albumin serving as a control. Phosphorylated Smad3 was detected by immunoprecipitation/Western blotting and confocal microscopy. Blocking studies using receptor for AGE siRNA and a specific inhibitor of Smad3 (SIS3) were performed in MMECs and in STZ-induced diabetic nephropathy in Tie2-Cre;Loxp-EGFP mice. RESULTS: Confocal microscopy and real-time PCR demonstrated that AGEs induced EndoMT in MMECs and in Tie2-Cre;Loxp-EGFP mice. Immunoprecipitation/Western blotting showed that Smad3 was activated by AGEs but was inhibited by SIS3 in MMECs and in STZ-induced diabetic nephropathy. Confocal microscopy and real-time PCR further demonstrated that SIS3 abrogated EndoMT, reduced renal fibrosis, and retarded progression of nephropathy. CONCLUSIONS: EndoMT is a novel pathway leading to early development of diabetic nephropathy. Blockade of EndoMT by SIS3 may provide a new strategy to retard the progression of diabetic nephropathy and other diabetes complications.


Asunto(s)
Diabetes Mellitus Experimental/fisiopatología , Nefropatías Diabéticas/prevención & control , Proteína smad3/genética , Animales , Diabetes Mellitus Experimental/prevención & control , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Proteínas de la Matriz Extracelular/fisiología , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/fisiología , Riñón/fisiología , Riñón/fisiopatología , Masculino , Mesodermo/fisiología , Ratones , Microscopía Confocal , Páncreas/irrigación sanguínea , Reacción en Cadena de la Polimerasa , Proteína-Lisina 6-Oxidasa/fisiología , ARN/genética , ARN/aislamiento & purificación , ARN Interferente Pequeño/genética , Receptor para Productos Finales de Glicación Avanzada , Receptor TIE-2/fisiología , Receptores Inmunológicos/fisiología , Albúmina Sérica/farmacología , Proteína Smad2/genética , Proteína smad3/antagonistas & inhibidores , Estreptozocina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA