Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Pharmacol Rep ; 76(3): 612-621, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38668812

RESUMEN

BACKGROUND: Podocytes have a remarkable ability to recover from injury; however, little is known about the recovery mechanisms involved in this process. We recently showed that formoterol, a long-acting ß2-adrenergic receptor (ß2-AR) agonist, induced mitochondrial biogenesis (MB) in podocytes and led to renoprotection in mice. However, it is not clear whether this effect was mediated by formoterol acting through the ß2-AR or if it occurred through "off-target" effects. METHODS: We genetically deleted the ß2-AR specifically in murine podocytes and used these mice to determine whether formoterol acting through the podocyte ß2-AR alone is sufficient for recovery of renal filtration function following injury. The podocyte-specific ß2-AR knockout mice (ß2-ARfl/fl/PodCre) were generated by crossing ß2-AR floxed mice with podocin Cre (B6.Cg-Tg(NPHS2-cre)295Lbh/J) mice. These mice were then subjected to both acute and chronic glomerular injury using nephrotoxic serum (NTS) and adriamycin (ADR), respectively. The extent of injury was evaluated by measuring albuminuria and histological and immunostaining analysis of the murine kidney sections. RESULTS: A similar level of injury was observed in ß2-AR knockout and control mice; however, the ß2-ARfl/fl/PodCre mice failed to recover in response to formoterol. Functional evaluation of the ß2-ARfl/fl/PodCre mice following injury plus formoterol showed similar albuminuria and glomerular injury to control mice that were not treated with formoterol. CONCLUSIONS: These results indicate that the podocyte ß2-AR is a critical component of the recovery mechanism and may serve as a novel therapeutic target for treating podocytopathies.


Asunto(s)
Doxorrubicina , Fumarato de Formoterol , Ratones Noqueados , Podocitos , Receptores Adrenérgicos beta 2 , Animales , Ratones , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Agonistas de Receptores Adrenérgicos beta 2/farmacología , Albuminuria/metabolismo , Doxorrubicina/farmacología , Doxorrubicina/toxicidad , Fumarato de Formoterol/farmacología , Ratones Endogámicos C57BL , Podocitos/metabolismo , Podocitos/efectos de los fármacos , Podocitos/patología , Receptores Adrenérgicos beta 2/metabolismo
2.
Am J Physiol Renal Physiol ; 326(1): F20-F29, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37916289

RESUMEN

We have previously shown that the long-acting ß2-adrenergic receptor (ß2-AR) agonist formoterol induced recovery from acute kidney injury in mice. To determine whether formoterol protected against diabetic nephropathy, the most common cause of end-stage kidney disease (ESKD), we used a high-fat diet (HFD), a murine type 2 diabetes model, and streptozotocin, a murine type 1 diabetes model. Following formoterol treatment, there was a marked recovery from and reversal of diabetic nephropathy in HFD mice compared with those treated with vehicle alone at the ultrastructural, histological, and functional levels. Similar results were seen after formoterol treatment in mice receiving streptozotocin. To investigate effects in humans, we performed a competing risk regression analysis with death as a competing risk to examine the association between Veterans with chronic kidney disease (CKD) and chronic obstructive pulmonary disease (COPD), who use ß2-AR agonists, and Veterans with CKD but no COPD, and progression to ESKD in a large national cohort of Veterans with stage 4 CKD between 2011 and 2013. Veterans were followed until 2016 or death. ESKD was defined as the initiation of dialysis and/or receipt of kidney transplant. We found that COPD was associated with a 25.6% reduction in progression from stage 4 CKD to ESKD compared with no COPD after adjusting for age, diabetes, sex, race-ethnicity, comorbidities, and medication use. Sensitivity analysis showed a 33.2% reduction in ESKD in Veterans with COPD taking long-acting formoterol and a 20.8% reduction in ESKD in Veterans taking other ß2-AR agonists compared with those with no COPD. These data indicate that ß2-AR agonists, especially formoterol, could be a treatment for diabetic nephropathy and perhaps other forms of CKD.NEW & NOTEWORTHY Diabetic nephropathy is the most common cause of ESKD. Formoterol, a long-acting ß2-adrenergic receptor (ß2-AR) agonist, reversed diabetic nephropathy in murine models of type 1 and 2 diabetes. In humans, there was an association with protection from progression of CKD in patients with COPD, by means of ß2-AR agonist intake, compared with those without COPD. These data indicate that ß2-AR agonists, especially formoterol, could be a new treatment for diabetic nephropathy and other forms of CKD.


Asunto(s)
Diabetes Mellitus Tipo 1 , Diabetes Mellitus Tipo 2 , Nefropatías Diabéticas , Fallo Renal Crónico , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Animales , Ratones , Nefropatías Diabéticas/tratamiento farmacológico , Agonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Estreptozocina , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Fumarato de Formoterol/uso terapéutico , Fallo Renal Crónico/tratamiento farmacológico , Fallo Renal Crónico/etiología , Receptores Adrenérgicos/uso terapéutico
3.
J Biol Chem ; 297(3): 101079, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34391780

RESUMEN

Phosphorylation (activation) and dephosphorylation (deactivation) of the slit diaphragm proteins NEPHRIN and NEPH1 are critical for maintaining the kidney epithelial podocyte actin cytoskeleton and, therefore, proper glomerular filtration. However, the mechanisms underlying these events remain largely unknown. Here we show that NEPHRIN and NEPH1 are novel receptor proteins for hepatocyte growth factor (HGF) and can be phosphorylated independently of the mesenchymal epithelial transition receptor in a ligand-dependent fashion through engagement of their extracellular domains by HGF. Furthermore, we demonstrate SH2 domain-containing protein tyrosine phosphatase-2-dependent dephosphorylation of these proteins. To establish HGF as a ligand, purified baculovirus-expressed NEPHRIN and NEPH1 recombinant proteins were used in surface plasma resonance binding experiments. We report high-affinity interactions of NEPHRIN and NEPH1 with HGF, although NEPHRIN binding was 20-fold higher than that of NEPH1. In addition, using molecular modeling we constructed peptides that were used to map specific HGF-binding regions in the extracellular domains of NEPHRIN and NEPH1. Finally, using an in vitro model of cultured podocytes and an ex vivo model of Drosophila nephrocytes, as well as chemically induced injury models, we demonstrated that HGF-induced phosphorylation of NEPHRIN and NEPH1 is centrally involved in podocyte repair. Taken together, this is the first study demonstrating a receptor-based function for NEPHRIN and NEPH1. This has important biological and clinical implications for the repair of injured podocytes and the maintenance of podocyte integrity.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Línea Celular , Tasa de Filtración Glomerular/fisiología , Factor de Crecimiento de Hepatocito/fisiología , Humanos , Uniones Intercelulares/metabolismo , Riñón/patología , Glomérulos Renales/metabolismo , Proteínas de la Membrana/genética , Ratones , Péptidos/metabolismo , Fosforilación , Podocitos/metabolismo , Unión Proteica/fisiología , Transducción de Señal/fisiología
4.
Cells ; 10(6)2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-34073294

RESUMEN

Unconventional myosins, linked to deafness, are also proposed to play a role in retinal cell physiology. However, their direct role in photoreceptor function remains unclear. We demonstrate that systemic loss of the unconventional myosin MYO1C in mice, specifically causes rhodopsin mislocalization, leading to impaired visual function. Electroretinogram analysis of Myo1c knockout (Myo1c-KO) mice showed a progressive loss of photoreceptor function. Immunohistochemistry and binding assays demonstrated MYO1C localization to photoreceptor inner and outer segments (OS) and identified a direct interaction of rhodopsin with MYO1C. In Myo1c-KO retinas, rhodopsin mislocalized to rod inner segments (IS) and cell bodies, while cone opsins in OS showed punctate staining. In aged mice, the histological and ultrastructural examination of the phenotype of Myo1c-KO retinas showed progressively shorter photoreceptor OS. These results demonstrate that MYO1C is important for rhodopsin localization to the photoreceptor OS, and for normal visual function.


Asunto(s)
Proteínas del Ojo/metabolismo , Células Fotorreceptoras/metabolismo , Retina/metabolismo , Rodopsina/metabolismo , Animales , Dineínas/genética , Electrorretinografía/métodos , Ratones , Fenotipo , Rodopsina/genética
5.
Am J Physiol Gastrointest Liver Physiol ; 320(6): G1044-G1053, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33908271

RESUMEN

Myosin 1c (Myo1c) is an unconventional myosin that modulates signaling pathways involved in tissue injury and repair. In this study, we observed that Myo1c expression is significantly upregulated in human chronic liver disease such as nonalcoholic steatohepatitis (NASH) and in animal models of liver fibrosis. High throughput data from the GEO-database identified similar Myo1c upregulation in mice and human liver fibrosis. Notably, transforming growth factor-ß1 (TGF-ß1) stimulation to hepatic stellate cells (HSCs), the liver pericyte and key cell type responsible for the deposition of extracellular matrix, upregulates Myo1c expression, whereas genetic depletion or pharmacological inhibition of Myo1c blunted TGF-ß-induced fibrogenic responses, resulting in repression of α-smooth muscle actin (α-SMA) and collagen type I α 1 chain (Col1α1) mRNA. Myo1c deletion also decreased fibrogenic processes such as cell proliferation, wound healing response, and contractility when compared with vehicle-treated HSCs. Importantly, phosphorylation of mothers against decapentaplegic homolog 2 (SMAD2) and mothers against decapentaplegic homolog 3 (SMAD3) were significantly blunted upon Myo1c inhibition in GRX cells as well as Myo1c knockout (Myo1c-KO) mouse embryonic fibroblasts (MEFs) upon TGF-ß stimulation. Using the genetic Myo1c-KO mice, we confirmed that Myo1c is critical for fibrogenesis, as Myo1c-KO mice were resistant to carbon tetrachloride (CCl4)-induced liver fibrosis. Histological and immunostaining analysis of liver sections showed that deposition of collagen fibers and α-SMA expression were significantly reduced in Myo1c-KO mice upon liver injury. Collectively, these results demonstrate that Myo1c mediates hepatic fibrogenesis by modulating TGF-ß signaling and suggest that inhibiting this process may have clinical application in treating liver fibrosis.NEW & NOTEWORTHY The incidences of liver fibrosis are growing at a rapid pace and have become one of the leading causes of end-stage liver disease. Although TGF-ß1 is known to play a prominent role in transforming cells to produce excessive extracellular matrix that lead to hepatic fibrosis, the therapies targeting TGF-ß1 have achieved very limited clinical impact. This study highlights motor protein myosin-1c-mediated mechanisms that serve as novel regulators of TGF-ß1 signaling and fibrosis.


Asunto(s)
Fibroblastos/metabolismo , Cirrosis Hepática/metabolismo , Hígado/metabolismo , Miosina Tipo I/metabolismo , Animales , Cadena alfa 1 del Colágeno Tipo I , Fibroblastos/patología , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Hígado/patología , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Ratones , Miosina Tipo I/genética , Fosforilación , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo
6.
Genes (Basel) ; 11(2)2020 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-32045989

RESUMEN

Sphingolipids are bioactive lipids associated with cellular membranes and plasma lipoproteins, and their synthesis and degradation are tightly regulated. We have previously determined that low plasma concentrations of certain ceramide species predict the development of nephropathy in diabetes patients with normal albumin excretion rates at baseline. Herein, we tested the hypothesis that altering the sphingolipid content of circulating lipoproteins can alter the metabolic and signaling pathways in podocytes, whose dysfunction leads to an impairment of glomerular filtration. Cultured human podocytes were treated with lipoproteins from healthy subjects enriched in vitro with C16 ceramide, or D-erythro 2-hydroxy C16 ceramide, a ceramide naturally found in skin. The RNA-Seq data demonstrated differential expression of genes regulating sphingolipid metabolism, sphingolipid signaling, and mTOR signaling pathways. A multiplex analysis of mTOR signaling pathway intermediates showed that the majority (eight) of the pathway phosphorylated proteins measured (eleven) were significantly downregulated in response to C16 ceramide-enriched HDL2 compared to HDL2 alone and hydroxy ceramide-enriched HDL2. In contrast, C16 ceramide-enriched HDL3 upregulated the phosphorylation of four intermediates in the mTOR pathway. These findings highlight a possible role for lipoprotein-associated sphingolipids in regulating metabolic and signaling pathways in podocytes and could lead to novel therapeutic targets in glomerular kidney diseases.


Asunto(s)
Ceramidas/metabolismo , Lipoproteínas/farmacología , Podocitos/metabolismo , Esfingolípidos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Transcriptoma/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Isótopos de Carbono , Línea Celular , Ceramidas/genética , HDL-Colesterol/farmacología , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/genética , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Fosforilación , Podocitos/efectos de los fármacos , RNA-Seq , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Esfingolípidos/genética , Serina-Treonina Quinasas TOR/genética , Transcriptoma/efectos de los fármacos
7.
Nat Commun ; 10(1): 4509, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31586055

RESUMEN

Fibroblast to myofibroblast differentiation is crucial for the initial healing response but excessive myofibroblast activation leads to pathological fibrosis. Therefore, it is imperative to understand the mechanisms underlying myofibroblast formation. Here we report that mitochondrial calcium (mCa2+) signaling is a regulatory mechanism in myofibroblast differentiation and fibrosis. We demonstrate that fibrotic signaling alters gating of the mitochondrial calcium uniporter (mtCU) in a MICU1-dependent fashion to reduce mCa2+ uptake and induce coordinated changes in metabolism, i.e., increased glycolysis feeding anabolic pathways and glutaminolysis yielding increased α-ketoglutarate (αKG) bioavailability. mCa2+-dependent metabolic reprogramming leads to the activation of αKG-dependent histone demethylases, enhancing chromatin accessibility in loci specific to the myofibroblast gene program, resulting in differentiation. Our results uncover an important role for the mtCU beyond metabolic regulation and cell death and demonstrate that mCa2+ signaling regulates the epigenome to influence cellular differentiation.


Asunto(s)
Señalización del Calcio/fisiología , Diferenciación Celular/genética , Epigénesis Genética/fisiología , Infarto del Miocardio/patología , Miofibroblastos/fisiología , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Metilación de ADN/fisiología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Epigenoma , Femenino , Fibrosis , Glucólisis/fisiología , Humanos , Ácidos Cetoglutáricos/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/etiología , Miocardio/citología , Miocardio/patología , Cultivo Primario de Células
8.
Kidney Int ; 96(4): 883-889, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31472902

RESUMEN

Steroid-resistant nephrotic syndrome is a frequent cause of chronic kidney disease almost inevitably progressing to end-stage renal disease. More than 58 monogenic causes of SRNS have been discovered and majority of known steroid-resistant nephrotic syndrome causing genes are predominantly expressed in glomerular podocytes, placing them at the center of disease pathogenesis. Herein, we describe two unrelated families with steroid-resistant nephrotic syndrome with homozygous mutations in the KIRREL1 gene. One mutation showed high frequency in the European population (minor allele frequency 0.0011) and this patient achieved complete remission following treatment, but later progressed to chronic kidney disease. We found that mutant KIRREL1 proteins failed to localize to the podocyte cell membrane, indicating defective trafficking and impaired podocytes function. Thus, the KIRREL1 gene product has an important role in modulating the integrity of the slit diaphragm and maintaining glomerular filtration function.


Asunto(s)
Resistencia a Medicamentos/genética , Glucocorticoides/farmacología , Proteínas de la Membrana/genética , Síndrome Nefrótico/genética , Insuficiencia Renal Crónica/genética , Adolescente , Edad de Inicio , Línea Celular , Niño , Preescolar , Consanguinidad , Análisis Mutacional de ADN , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Frecuencia de los Genes , Membrana Basal Glomerular/patología , Membrana Basal Glomerular/ultraestructura , Glucocorticoides/uso terapéutico , Homocigoto , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Microscopía Electrónica de Transmisión , Mutación , Síndrome Nefrótico/tratamiento farmacológico , Síndrome Nefrótico/patología , Linaje , Podocitos , Insuficiencia Renal Crónica/patología , Secuenciación del Exoma
9.
Kidney Int ; 96(3): 656-673, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31262488

RESUMEN

Podocytes have limited ability to recover from injury. Here, we demonstrate that increased mitochondrial biogenesis, to meet the metabolic and energy demand of a cell, accelerates podocyte recovery from injury. Analysis of events induced during podocyte injury and recovery showed marked upregulation of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), a transcriptional co-activator of mitochondrial biogenesis, and key components of the mitochondrial electron transport chain. To evaluate our hypothesis that increasing mitochondrial biogenesis enhanced podocyte recovery from injury, we treated injured podocytes with formoterol, a potent, specific, and long-acting ß2-adrenergic receptor agonist that induces mitochondrial biogenesis in vitro and in vivo. Formoterol increased mitochondrial biogenesis and restored mitochondrial morphology and the injury-induced changes to the organization of the actin cytoskeleton in podocytes. Importantly, ß2-adrenergic receptors were found to be present on podocyte membranes. Their knockdown attenuated formoterol-induced mitochondrial biogenesis. To determine the potential clinical relevance of these findings, mouse models of acute nephrotoxic serum nephritis and chronic (Adriamycin [doxorubicin]) glomerulopathy were used. Mice were treated with formoterol post-injury when glomerular dysfunction was established. Strikingly, formoterol accelerated the recovery of glomerular function by reducing proteinuria and ameliorating kidney pathology. Furthermore, formoterol treatment reduced cellular apoptosis and increased the expression of the mitochondrial biogenesis marker PGC-1α and multiple electron transport chain proteins. Thus, our results support ß2-adrenergic receptors as novel therapeutic targets and formoterol as a therapeutic compound for treating podocytopathies.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2/farmacología , Fumarato de Formoterol/farmacología , Glomerulonefritis/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Podocitos/efectos de los fármacos , Agonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Doxorrubicina/toxicidad , Fumarato de Formoterol/uso terapéutico , Técnicas de Silenciamiento del Gen , Glomerulonefritis/inducido químicamente , Glomerulonefritis/patología , Humanos , Ratones , Mitocondrias/metabolismo , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Podocitos/citología , Podocitos/patología , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal
10.
Kidney Int ; 96(1): 139-158, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31097328

RESUMEN

Transforming growth factor-ß (TGF-ß) is known to play a critical role in the pathogenesis of many progressive podocyte diseases. However, the molecular mechanisms regulating TGF-ß signaling in podocytes remain unclear. Using a podocyte-specific myosin (Myo)1c knockout, we demonstrate whether Myo1c is critical for TGF-ß-signaling in podocyte disease pathogenesis. Specifically, podocyte-specific Myo1c knockout mice were resistant to fibrotic injury induced by Adriamycin or nephrotoxic serum. Further, loss of Myo1c also protected from injury in the TGF-ß-dependent unilateral ureteral obstruction mouse model of renal interstitial fibrosis. Mechanistic analyses showed that loss of Myo1c significantly blunted TGF-ß signaling through downregulation of canonical and non-canonical TGF-ß pathways. Interestingly, nuclear rather than the cytoplasmic Myo1c was found to play a central role in controlling TGF-ß signaling through transcriptional regulation. Differential expression analysis of nuclear Myo1c-associated gene promoters showed that nuclear Myo1c targeted the TGF-ß responsive gene growth differentiation factor (GDF)-15 and directly bound to the GDF-15 promoter. Importantly, GDF15 was found to be involved in podocyte pathogenesis, where GDF15 was upregulated in glomeruli of patients with focal segmental glomerulosclerosis. Thus, Myo1c-mediated regulation of TGF-ß-responsive genes is central to the pathogenesis of podocyte injury. Hence, inhibiting this process may have clinical application in treating podocytopathies.


Asunto(s)
Factor 15 de Diferenciación de Crecimiento/genética , Enfermedades Renales/patología , Miosina Tipo I/metabolismo , Podocitos/patología , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Modelos Animales de Enfermedad , Doxorrubicina/toxicidad , Femenino , Fibrosis , Regulación de la Expresión Génica , Humanos , Enfermedades Renales/inducido químicamente , Masculino , Ratones , Ratones Noqueados , Miosina Tipo I/genética , Podocitos/efectos de los fármacos , Regiones Promotoras Genéticas , Transcripción Genética
11.
J Biol Chem ; 294(26): 10104-10119, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31073028

RESUMEN

Although the slit diaphragm proteins in podocytes are uniquely organized to maintain glomerular filtration assembly and function, little is known about the underlying mechanisms that participate in trafficking these proteins to the correct location for development and homeostasis. Identifying these mechanisms will likely provide novel targets for therapeutic intervention to preserve podocyte function following glomerular injury. Analysis of structural variation in cases of human nephrotic syndrome identified rare heterozygous deletions of EXOC4 in two patients. This suggested that disruption of the highly-conserved eight-protein exocyst trafficking complex could have a role in podocyte dysfunction. Indeed, mRNA profiling of injured podocytes identified significant exocyst down-regulation. To test the hypothesis that the exocyst is centrally involved in podocyte development/function, we generated homozygous podocyte-specific Exoc5 (a central exocyst component that interacts with Exoc4) knockout mice that showed massive proteinuria and died within 4 weeks of birth. Histological and ultrastructural analysis of these mice showed severe glomerular defects with increased fibrosis, proteinaceous casts, effaced podocytes, and loss of the slit diaphragm. Immunofluorescence analysis revealed that Neph1 and Nephrin, major slit diaphragm constituents, were mislocalized and/or lost. mRNA profiling of Exoc5 knockdown podocytes showed that vesicular trafficking was the most affected cellular event. Mapping of signaling pathways and Western blot analysis revealed significant up-regulation of the mitogen-activated protein kinase and transforming growth factor-ß pathways in Exoc5 knockdown podocytes and in the glomeruli of podocyte-specific Exoc5 KO mice. Based on these data, we propose that exocyst-based mechanisms regulate Neph1 and Nephrin signaling and trafficking, and thus podocyte development and function.


Asunto(s)
Eliminación de Gen , Glomérulos Renales/patología , Síndrome Nefrótico/patología , Podocitos/patología , Proteínas de Transporte Vesicular/fisiología , Animales , Apoptosis , Movimiento Celular , Exocitosis , Humanos , Glomérulos Renales/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome Nefrótico/genética , Fosforilación , Podocitos/metabolismo , Transporte de Proteínas , Proteinuria/etiología , Proteinuria/patología , Transducción de Señal
12.
Nephrology (Carlton) ; 24(5): 497-503, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30848004

RESUMEN

Beta2-adrenergic receptor (ß2 -AR) is a G-protein-coupled adrenergic receptor family member, whose clinical significance has been extensively investigated in lung, cardiovascular and muscular diseases, but its role in kidney biology remains understudied. In this review, we discuss some of the recent studies, where the effect of agonist/antagonist-mediated activation/inhibition of ß2 -AR on disease pathogenesis process was studied, and highlighted the role of ß2 -AR in kidney biology. The expression of ß2 -AR has been noted in many kidney subunits including proximal tubules, glomeruli and podocytes. In vivo studies have shown that in cultured proximal tubules ß2 -AR is involved in Na-ATPase activity and transcellular Na-transport through protein kinase-C activation; whereas in cultured podocytes, it was associated with depolarization of the membrane. The animal studies further revealed that ß2 -AR activation by short-acting ß2 agonists attenuated monocyte activation, pro-inflammatory and pro-fibrotic responses through ß-arrestin2 dependent NF-kB inactivation in diabetic kidney disease; in contrast, activation by long-acting ß2 agonists restored mitochondrial and renal function in the acute kidney injury mice models through PGC-1α dependent mitochondrial biogenesis. In conclusion, the activation of ß2 -AR may present a rapidly developing therapeutic target for renal diseases.


Asunto(s)
Lesión Renal Aguda/metabolismo , Nefropatías Diabéticas/metabolismo , Riñón/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Agonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Animales , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Riñón/fisiopatología , Receptores Adrenérgicos beta 2/efectos de los fármacos , Fármacos Renales/uso terapéutico , Transducción de Señal
13.
Kidney Int ; 95(3): 708-716, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30709661

RESUMEN

Definitive diagnosis of glomerular disease requires a kidney biopsy, an invasive procedure that may not be safe or feasible to perform in all patients. We developed a noninvasive, accurate, and economical diagnostic assay with easy commercial adaptability to detect recurrent focal segmental glomerulosclerosis (rFSGS) after kidney transplant. Since FSGS involves podocyte damage and death, our approach involved mRNA profiling of cultured podocytes treated with plasma from patients with rFSGS to identify upregulated genes involved in podocyte damage. For concept validation, three upregulated pro-apoptotic candidate genes (IL1ß, BMF, and IGFBP3) were selected, and their promoter regions were cloned into a luciferase-based reporter vector and transfected into podocytes to generate stable podocyte cell lines. Strikingly, when exposed to rFSGS patient plasma, these cell lines showed increased reporter activity; in contrast, no reporter activity was noted with plasma from patients with non-recurrent FSGS or membranous nephropathy. Area under the receiver operating characteristics curves (AUCs) for models discriminating between rFSGS and other nephropathies (non-recurrent FSGS and membranous nephropathy) and between rFSGS and non-recurrent FSGS ranged from 0.81 to 0.86, respectively. Estimated sensitivities and specificities for the diagnosis of rFSGS were greater than 80% for the IL1ß and BMF cell lines, and were slightly lower for the IGFBP3 cell line. Importantly, the novel approach outlined here for the diagnosis of rFSGS is widely applicable to the design of sensitive and specific diagnostic/prognostic assays for other glomerular diseases.


Asunto(s)
Bioensayo/métodos , Glomeruloesclerosis Focal y Segmentaria/diagnóstico , Podocitos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular , Diagnóstico Diferencial , Perfilación de la Expresión Génica , Genes Reporteros , Glomeruloesclerosis Focal y Segmentaria/sangre , Glomeruloesclerosis Focal y Segmentaria/complicaciones , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Fallo Renal Crónico/etiología , Fallo Renal Crónico/cirugía , Trasplante de Riñón , Luciferasas/genética , Plasma/metabolismo , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo , RNA-Seq , Curva ROC , Recurrencia
14.
Int J Mol Sci ; 21(1)2019 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-31906131

RESUMEN

Podocytes have a unique structure that supports glomerular filtration function, and many glomerular diseases result in loss of this structure, leading to podocyte dysfunction and ESRD (end stage renal disease). These structural and functional changes involve a complex set of molecular and cellular mechanisms that remain poorly understood. To understand the molecular signature of podocyte injury, we performed transcriptome analysis of cultured human podocytes injured either with PAN (puromycin aminonucleoside) or doxorubicin/adriamycin (ADR). The pathway analysis through DE (differential expression) and gene-enrichment analysis of the injured podocytes showed Tumor protein p53 (P53) as one of the major signaling pathways that was significantly upregulated upon podocyte injury. Accordingly, P53 expression was also up-regulated in the glomeruli of nephrotoxic serum (NTS) and ADR-injured mice. To further confirm these observations, cultured podocytes were treated with the P53 inhibitor pifithrin-α, which showed significant protection from ADR-induced actin cytoskeleton damage. In conclusion, signaling pathways that are involved in podocyte pathogenesis and can be therapeutically targeted were identified by high-throughput transcriptomic analysis of injured podocytes.


Asunto(s)
Doxorrubicina/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Enfermedades Renales/metabolismo , Podocitos/metabolismo , Puromicina Aminonucleósido/efectos adversos , Transducción de Señal/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Animales , Doxorrubicina/farmacología , Humanos , Enfermedades Renales/inducido químicamente , Enfermedades Renales/patología , Ratones , Fosforilación/efectos de los fármacos , Podocitos/patología , Puromicina Aminonucleósido/farmacología
15.
Kidney Int Rep ; 3(6): 1443-1453, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30426109

RESUMEN

INTRODUCTION: Tubular dysfunction is characteristic of Dent's disease; however, focal segmental glomerulosclerosis (FSGS) can also be present. Glomerulosclerosis could be secondary to tubular injury, but it remains uncertain whether the CLCN5 gene, which encodes an endosomal chloride and/or hydrogen exchanger, plays a role in podocyte biology. Here, we implicate a role for CLCN5 in podocyte function and pathophysiology. METHODS: Whole exome capture and sequencing of the proband and 5 maternally-related family members was conducted to identify X-linked mutations associated with biopsy-proven FSGS. Human podocyte cultures were used to characterize the mutant phenotype on podocyte function. RESULTS: We identified a novel mutation (L521F) in CLCN5 in 2 members of a Hispanic family who presented with a histologic diagnosis of FSGS and low-molecular-weight proteinuria without hypercalciuria. Presence of CLCN5 was confirmed in cultured human podocytes. Podocytes transfected with the wild-type or the mutant (L521F) CLCN5 constructs showed differential localization. CLCN5 knockdown in podocytes resulted in defective transferrin endocytosis and was associated with decreased cell proliferation and increased cell migration, which are hallmarks of podocyte injury. CONCLUSIONS: The CLCN5 mutation, which causes Dent's disease, may be associated with FSGS without hyercalcuria and nepthrolithiasis. The present findings supported the hypothesis that CLCN5 participates in protein trafficking in podocytes and plays a critical role in organizing the components of the podocyte slit diaphragm to help maintain normal cell physiology and a functional filtration barrier. In addition to tubular dysfunction, mutations in CLCN5 may also lead to podocyte dysfunction, which results in a histologic picture of FSGS that may be a primary event and not a consequence of tubular damage.

16.
Am J Physiol Renal Physiol ; 314(2): F280-F292, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29046299

RESUMEN

Podocyte dysfunction and loss is an early event and a hallmark of proteinuric kidney diseases. A podocyte's normal function is maintained via its unique cellular architecture that relies on an intracellular network of filaments, including filamentous actin (F-actin) and microtubules, that provides mechanical support. Damage to this filamentous network leads to changes in cellular morphology and results in podocyte injury, dysfunction, and death. Conversely, stabilization of this network protects podocytes and ameliorates proteinuria. This suggests that stabilization of podocyte architecture via its filamentous network could be a key therapeutic strategy for proteinuric kidney diseases. However, development of podocyte-directed therapeutics, especially those that target the cell's filamentous network, is still lacking, partly because of unavailability of appropriate cellular assays for use in a drug discovery environment. Here, we describe a new high-content screening-based methodology and its implementation on podocytes to identify paullone derivatives as a novel group of podocyte-protective compounds. We find that three compounds, i.e., kenpaullone, 1-azakenpaullone, and alsterpaullone, dose dependently protect podocytes from puromycin aminonucleoside (PAN)-mediated injury in vitro by reducing PAN-induced changes in both the filamentous actin and microtubules, with alsterpaullone providing maximal protection. Mechanistic studies further show that alsterpaullone suppressed PAN-induced activation of signaling downstream of GSK3ß and p38 mitogen-activated protein kinase. In vivo it reduced ADR-induced glomerular injury in a zebrafish model. Together, these results identify paullone derivatives as novel podocyte-protective agents for future therapeutic development.


Asunto(s)
Benzazepinas/farmacología , Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento , Indoles/farmacología , Podocitos/efectos de los fármacos , Sustancias Protectoras/farmacología , Fármacos Renales/farmacología , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Doxorrubicina , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Enfermedades Renales/inducido químicamente , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Ratones , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Microtúbulos/patología , Podocitos/metabolismo , Podocitos/patología , Transducción de Señal/efectos de los fármacos , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
17.
Sci Rep ; 7(1): 12047, 2017 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-28935902

RESUMEN

Targeting protein-protein interaction (PPI) is rapidly becoming an attractive alternative for drug development. While drug development commonly involves inhibiting a PPI, in this study, we show that stabilizing PPI may also be therapeutically beneficial. Junctional proteins Neph1 and ZO-1 and their interaction is an important determinant of the structural integrity of slit diaphragm, which is a critical component of kidney's filtration system. Since injury induces loss of this interaction, we hypothesized that strengthening this interaction may protect kidney's filtration barrier and preserve kidney function. In this study, Neph1-ZO-1 structural complex was screened for the presence of small druggable pockets formed from contributions from both proteins. One such pocket was identified and screened using a small molecule library. Isodesmosine (ISD) a rare naturally occurring amino acid and a biomarker for pulmonary arterial hypertension was selected as the best candidate and to establish the proof of concept, its ability to enhance Neph1-CD and ZO-1 binding was tested. Results from biochemical binding analysis showed that ISD enhanced Neph1 and ZO-1 interaction under in vitro and in vivo conditions. Importantly, ISD treated podocytes were resistant to injury-induced loss of transepithelial permeability. Finally, mouse and zebrafish studies show that ISD protects from injury-induced renal damage.


Asunto(s)
Isodesmosina/farmacología , Proteínas de la Membrana/metabolismo , Podocitos/efectos de los fármacos , Proteína de la Zonula Occludens-1/metabolismo , Animales , Células Cultivadas , Tasa de Filtración Glomerular/efectos de los fármacos , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/fisiopatología , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Simulación del Acoplamiento Molecular , Podocitos/metabolismo , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Pez Cebra , Proteína de la Zonula Occludens-1/química , Proteína de la Zonula Occludens-1/genética
18.
J Am Soc Nephrol ; 28(7): 2119-2132, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28202497

RESUMEN

Aminopeptidase A (APA) is expressed in glomerular podocytes and tubular epithelia and metabolizes angiotensin II (AngII), a peptide known to promote glomerulosclerosis. In this study, we tested whether APA expression changes in response to progressive nephron loss or whether APA exerts a protective role against glomerular damage and during AngII-mediated hypertensive kidney injury. At advanced stages of FSGS, fawn-hooded hypertensive rat kidneys exhibited distinctly increased APA staining in areas of intact glomerular capillary loops. Moreover, BALB/c APA-knockout (KO) mice injected with a nephrotoxic serum showed persistent glomerular hyalinosis and albuminuria 96 hours after injection, whereas wild-type controls achieved virtually full recovery. We then tested the effect of 4-week infusion of AngII (400 ng/kg per minute) in APA-KO and wild-type mice. Although we observed no significant difference in achieved systolic BP, AngII-treated APA-KO mice developed a significant rise in albuminuria not observed in AngII-treated wild-type mice along with increased segmental and global sclerosis and/or collapse of juxtamedullary glomeruli, microcystic tubular dilation, and tubulointerstitial fibrosis. In parallel, AngII treatment significantly increased the kidney AngII content and attenuated the expression of podocyte nephrin in APA-KO mice but not in wild-type controls. These data show that deficiency of APA increases susceptibility to glomerular injury in BALB/c mice. The augmented AngII-mediated kidney injury observed in association with increased intrarenal AngII accumulation in the absence of APA suggests a protective metabolizing role of APA in AngII-mediated glomerular diseases.


Asunto(s)
Glutamil Aminopeptidasa/deficiencia , Enfermedades Renales/enzimología , Enfermedades Renales/etiología , Glomérulos Renales , Animales , Susceptibilidad a Enfermedades , Masculino , Ratones , Ratones Noqueados , Ratas
19.
Mol Cell Biol ; 36(11): 1639-54, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27044863

RESUMEN

The Myo1c motor functions as a cargo transporter supporting various cellular events, including vesicular trafficking, cell migration, and stereociliary movements of hair cells. Although its partial crystal structures were recently described, the structural details of its interaction with cargo proteins remain unknown. This study presents the first structural demonstration of a cargo protein, Neph1, attached to Myo1c, providing novel insights into the role of Myo1c in intracellular movements of this critical slit diaphragm protein. Using small angle X-ray scattering studies, models of predominant solution conformation of unliganded full-length Myo1c and Myo1c bound to Neph1 were constructed. The resulting structures show an extended S-shaped Myo1c with Neph1 attached to its C-terminal tail. Importantly, binding of Neph1 did not induce a significant shape change in Myo1c, indicating this as a spontaneous process or event. Analysis of interaction surfaces led to the identification of a critical residue in Neph1 involved in binding to Myo1c. Indeed, a point mutant from this site abolished interaction between Neph1 and Myo1c when tested in the in vitro and in live-cell binding assays. Live-cell imaging, including fluorescence recovery after photobleaching, provided further support for the role of Myo1c in intracellular vesicular movement of Neph1 and its turnover at the membrane.


Asunto(s)
Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Miosina Tipo I/química , Miosina Tipo I/metabolismo , Sitios de Unión , Células Cultivadas , Humanos , Proteínas de la Membrana/genética , Modelos Moleculares , Podocitos/metabolismo , Podocitos/ultraestructura , Mutación Puntual , Unión Proteica , Transporte de Proteínas , Dispersión del Ángulo Pequeño , Difracción de Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA