Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Kidney Int ; 103(1): 144-155, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36273656

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) involves the development and persistent growth of fluid filled kidney cysts. In a recent study, we showed that ADPKD kidney cyst epithelial cells can stimulate the proliferation and differentiation of peri-cystic myofibroblasts. Although dense myofibroblast populations are often found surrounding kidney cysts, their role in cyst enlargement or fibrosis in ADPKD is unclear. To clarify this, we examined the effect of myofibroblast depletion in the Pkd1RC/RC (RC/RC) mouse model of ADPKD. RC/RC;αSMAtk mice that use the ganciclovir-thymidine kinase system to selectively deplete α-smooth muscle actin expressing myofibroblasts were generated. Ganciclovir treatment for four weeks depleted myofibroblasts, reduced kidney fibrosis and preserved kidney function in these mice. Importantly, myofibroblast depletion significantly reduced cyst growth and cyst epithelial cell proliferation in RC/RC;αSMAtk mouse kidneys. Similar ganciclovir treatment did not alter cyst growth or fibrosis in wild-type or RC/RC littermates. In vitro, co-culture with myofibroblasts from the kidneys of patients with ADPKD increased 3D microcyst growth of human ADPKD cyst epithelial cells. Treatment with conditioned culture media from ADPKD kidney myofibroblasts increased microcyst growth and cell proliferation of ADPKD cyst epithelial cells. Further examination of ADPKD myofibroblast conditioned media showed high levels of protease inhibitors including PAI1, TIMP1 and 2, NGAL and TFPI-2, and treatment with recombinant PAI1 and TIMP1 increased ADPKD cyst epithelial cell proliferation in vitro. Thus, our findings show that myofibroblasts directly promote cyst epithelial cell proliferation, cyst growth and fibrosis in ADPKD kidneys, and their targeting could be a novel therapeutic strategy to treat PKD.


Asunto(s)
Quistes , Riñón Poliquístico Autosómico Dominante , Humanos , Ratones , Animales , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Miofibroblastos , Células Cultivadas , Riñón/patología , Proliferación Celular , Fibrosis , Quistes/tratamiento farmacológico , Quistes/patología , Células Epiteliales/patología
2.
Int J Mol Sci ; 23(14)2022 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-35886951

RESUMEN

Vasopressin type-2 receptor (V2R) is ectopically expressed and plays a pathogenic role in clear cell renal cell carcinoma (ccRCC) tumor cells. Here we examined how V2R signaling within human ccRCC tumor cells (Caki1 cells) stimulates stromal cancer-associated fibroblasts (CAFs). We found that cell culture conditioned media from Caki1 cells increased activation, migration, and proliferation of fibroblasts in vitro, which was inhibited by V2R gene silencing in Caki1 cells. Analysis of the conditioned media and mRNA of the V2R gene silenced and control Caki1 cells showed that V2R regulates the production of CAF-activating factors. Some of these factors were also found to be regulated by YAP in these Caki1 cells. YAP expression colocalized and correlated with V2R expression in ccRCC tumor tissue. V2R gene silencing or V2R antagonist significantly reduced YAP in Caki1 cells. Moreover, the V2R antagonist reduced YAP expression and myofibroblasts in mouse xenograft tumors. These results suggest that V2R plays an important role in secreting pro-fibrotic factors that stimulate fibroblast activation by a YAP-dependent mechanism in ccRCC tumors. Our results demonstrate a novel role for the V2R-YAP axis in the regulation of myofibroblasts in ccRCC and a potential therapeutic target.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinoma de Células Renales , Neoplasias Renales , Receptores de Vasopresinas , Animales , Antagonistas de los Receptores de Hormonas Antidiuréticas/farmacología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Medios de Cultivo Condicionados , Fibroblastos/metabolismo , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Ratones , Receptores de Vasopresinas/genética , Receptores de Vasopresinas/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/fisiología , Vasopresinas/genética , Vasopresinas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
FASEB J ; 34(10): 13597-13608, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32813289

RESUMEN

Acute kidney injury (AKI) is characterized by injury to the tubular epithelium that leads to the sudden loss of renal function. Proper tubular regeneration is essential to prevent progression to chronic kidney disease. In this study, we examined the role of FoxM1, a forkhead box family member transcription factor in tubular repair after AKI. Renal FoxM1 expression increased after renal ischemia/reperfusion (I/R)-induced AKI in mouse kidneys. Treatment with thiostrepton, a FoxM1 inhibitor, reduced FoxM1 regulated pro-proliferative factors and cell proliferation in vitro, and tubular regeneration in mouse kidneys after AKI. Glycogen synthase kinase-3 (GSK3) was found to be an upstream regulator of FoxM1 because GSK3 inhibition or renal tubular GSK3ß gene deletion significantly increased FoxM1 expression, and improved tubular repair and renal function. GSK3 inactivation increased ß-catenin, Cyclin D1, and c-Myc, and reduced cell cycle inhibitors p21 and p27. Importantly, thiostrepton treatment abolished the improved tubular repair in GSK3ß knockout mice following AKI. These results demonstrate that FoxM1 is important for renal tubular regeneration following AKI and that GSK3ß suppresses tubular repair by inhibiting FoxM1.


Asunto(s)
Lesión Renal Aguda/metabolismo , Proteína Forkhead Box M1/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Daño por Reperfusión/metabolismo , Animales , Línea Celular , Proliferación Celular , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Túbulos Renales/patología , Túbulos Renales/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regeneración
4.
J Am Soc Nephrol ; 31(8): 1697-1710, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32554753

RESUMEN

BACKGROUND: Fibrosis is a major cause of loss of renal function in autosomal dominant polycystic kidney disease (ADPKD). In this study, we examined whether vasopressin type-2 receptor (V2R) activity in cystic epithelial cells can stimulate interstitial myofibroblasts and fibrosis in ADPKD kidneys. METHODS: We treated Pkd1 gene knockout (Pkd1KO) mice with dDAVP, a V2R agonist, for 3 days and evaluated the effect on myofibroblast deposition of extracellular matrix (ECM). We also analyzed the effects of conditioned media from primary cultures of human ADPKD cystic epithelial cells on myofibroblast activation. Because secretion of the profibrotic connective tissue growth factor (CCN2) increased significantly in dDAVP-treated Pkd1KO mouse kidneys, we examined its role in V2R-dependent fibrosis in ADPKD as well as that of yes-associated protein (YAP). RESULTS: V2R stimulation using dDAVP increased the renal interstitial myofibroblast population and ECM deposition. Similarly, conditioned media from human ADPKD cystic epithelial cells increased myofibroblast activation in vitro, suggesting a paracrine mechanism. Renal collecting duct-specific gene deletion of CCN2 significantly reduced cyst growth and myofibroblasts in Pkd1KO mouse kidneys. We found that YAP regulates CCN2, and YAP inhibition or gene deletion reduces renal fibrosis in Pkd1KO mouse kidneys. Importantly, YAP inactivation blocks the dDAVP-induced increase in myofibroblasts in Pkd1KO kidneys. Further in vitro studies showed that V2R regulates YAP by an ERK1/2-dependent mechanism in human ADPKD cystic epithelial cells. CONCLUSIONS: Our results demonstrate a novel mechanism by which cystic epithelial cells stimulate myofibroblasts in the pericystic microenvironment, leading to fibrosis in ADPKD. The V2R-YAP-CCN2 cell signaling pathway may present a potential therapeutic target for fibrosis in ADPKD.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Factor de Crecimiento del Tejido Conjuntivo/fisiología , Riñón/patología , Miofibroblastos/fisiología , Riñón Poliquístico Autosómico Dominante/patología , Receptores de Vasopresinas/fisiología , Factores de Transcripción/fisiología , Animales , Desamino Arginina Vasopresina/farmacología , Matriz Extracelular/metabolismo , Fibrosis , Humanos , Ratones , Canales Catiónicos TRPP/fisiología
5.
Int J Mol Sci ; 22(19)2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34638957

RESUMEN

The tumor microenvironment plays a critical role in defining the growth and malignancy of solid tumors. Extracellular matrix (ECM) proteins such as collagen, vitronectin, and fibronectin are major components of the tumor microenvironment. Tumor growth-promoting reciprocal interaction between ECM and cytoplasmic proteins is regulated by the cell surface receptors called integrins. This study investigated the mechanism by which integrin ß1 promotes pancreatic tumor growth. In MIA PaCa-2 pancreatic cancer cell line, the loss of integrin ß1 protein reduced the ability of cells to proliferate in a 3D matrix and compromised the ability to form a focal adhesion complex. Decreased expression of integrin α5 was observed in KO cells, which resulted in impaired cell spreading and adhesion on vitronectin and fibronectin. Reduced expression of the integrin-associated protein, kindlin-2 was also recorded. The downregulation of kindlin-2 decreased the phosphorylation of Smad2/3 by reducing the expression of TGF-ß receptor 2. These results unravel a new mechanism of integrin ß1 in tumor growth by modifying the expression of kindlin-2 and TGF-ß receptor 2 signaling.


Asunto(s)
Carcinogénesis/metabolismo , Proliferación Celular/genética , Integrina beta1/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal/genética , Carcinogénesis/genética , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Integrina beta1/genética , Neoplasias Pancreáticas/patología , Transfección
6.
Am J Physiol Renal Physiol ; 317(2): F343-F360, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31091126

RESUMEN

Polycystic kidney disease (PKD) is characterized by slowly expanding renal cysts that damage the kidney, typically resulting in renal failure by the fifth decade. The most common cause of death in these patients, however, is cardiovascular disease. Expanding cysts in PKD induce chronic kidney injury that is accompanied by immune cell infiltration, including macrophages, which we and others have shown can promote disease progression in PKD mouse models. Here, we show that monocyte chemoattractant protein-1 [MCP-1/chemokine (C-C motif) ligand 2 (CCL2)] is responsible for the majority of monocyte chemoattractant activity produced by renal PKD cells from both mice and humans. To test whether the absence of MCP-1 lowers renal macrophage concentration and slows disease progression, we generated genetic knockout (KO) of MCP-1 in a mouse model of PKD [congenital polycystic kidney (cpk) mice]. Cpk mice are born with rapidly expanding renal cysts, accompanied by a decline in kidney function and death by postnatal day 21. Here, we report that KO of MCP-1 in these mice increased survival, with some mice living past 3 mo. Surprisingly, however, there was no significant difference in renal macrophage concentration, nor was there improvement in cystic disease or kidney function. Examination of mice revealed cardiac hypertrophy in cpk mice, and measurement of cardiac electrical activity via ECG revealed repolarization abnormalities. MCP-1 KO did not affect the number of cardiac macrophages, nor did it alleviate the cardiac aberrancies. However, MCP-1 KO did prevent the development of pulmonary edema, which occurred in cpk mice, and promoted decreased resting heart rate and increased heart rate variability in both cpk and noncystic mice. These data suggest that in this mouse model of PKD, MCP-1 altered cardiac/pulmonary function and promoted death outside of its role as a macrophage chemoattractant.


Asunto(s)
Arritmias Cardíacas/metabolismo , Cardiomegalia/metabolismo , Quimiocina CCL2/metabolismo , Riñón/metabolismo , Pulmón/metabolismo , Miocardio/metabolismo , Enfermedades Renales Poliquísticas/metabolismo , Edema Pulmonar/metabolismo , Animales , Arritmias Cardíacas/patología , Arritmias Cardíacas/fisiopatología , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Células Cultivadas , Quimiocina CCL2/deficiencia , Quimiocina CCL2/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis , Humanos , Mediadores de Inflamación/metabolismo , Riñón/patología , Riñón/fisiopatología , Pulmón/patología , Pulmón/fisiopatología , Macrófagos/metabolismo , Macrófagos/patología , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Enfermedades Renales Poliquísticas/patología , Enfermedades Renales Poliquísticas/fisiopatología , Edema Pulmonar/patología , Edema Pulmonar/fisiopatología , Edema Pulmonar/prevención & control , Factores de Tiempo
7.
J Am Soc Nephrol ; 27(6): 1604-7, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27026364

RESUMEN

The Kidney Tutored Research and Education for Kidney Students (TREKS) Program is a product of the American Society of Nephrology (ASN) Workforce Committee that seeks to connect medical and graduate students to nephrology. This program starts with a weeklong camp-like course introducing participants to renal physiology through classic and modern experiments. Next, each student is matched with a nephrology mentor at his or her home institution to foster a better understanding of a nephrology career. Lastly, the students are encouraged to participate in scholarly activities and attend the ASN Kidney Week. Now in its third year, with a total of 84 participants, survey data suggest early success of the program, with a self-reported 40% increased interest in nephrology fellowship and/or research careers. In addition, students give high ratings to the course components and mentorship pairings. Continued student tracking will be necessary to determine the long-term program effect.


Asunto(s)
Selección de Profesión , Nefrología/educación , Educación de Postgrado en Medicina , Femenino , Humanos , Masculino , Mentores , Sociedades Médicas , Estados Unidos
8.
Am J Physiol Renal Physiol ; 308(6): F650-60, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25608967

RESUMEN

In mammals, glycogen synthase kinase (GSK)3 comprises GSK3α and GSK3ß isoforms. GSK3ß has been shown to play a role in the ability of kidneys to concentrate urine by regulating vasopressin-mediated water permeability of collecting ducts, whereas the role of GSK3α has yet to be discerned. To investigate the role of GSK3α in urine concentration, we compared GSK3α knockout (GSK3αKO) mice with wild-type (WT) littermates. Under normal conditions, GSK3αKO mice had higher water intake and urine output. GSK3αKO mice also showed reduced urine osmolality and aquaporin-2 levels but higher urinary vasopressin. When water deprived, they failed to concentrate their urine to the same level as WT littermates. The addition of 1-desamino-8-d-arginine vasopressin to isolated inner medullary collecting ducts increased the cAMP response in WT mice, but this response was reduced in GSK3αKO mice, suggesting reduced responsiveness to vasopressin. Gene silencing of GSK3α in mpkCCD cells also reduced forskolin-induced aquaporin-2 expression. When treated with LiCl, an isoform nonselective inhibitor of GSK3 and known inducer of polyuria, WT mice developed significant polyuria within 6 days. However, in GSK3αKO mice, the polyuric response was markedly reduced. This study demonstrates, for the first time, that GSK3α could play a crucial role in renal urine concentration and suggest that GSK3α might be one of the initial targets of Li(+) in LiCl-induced nephrogenic diabetes insipidus.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Túbulos Renales Colectores/enzimología , Orina/fisiología , Animales , Acuaporina 2/metabolismo , Silenciador del Gen , Glucógeno Sintasa Quinasa 3/genética , Cloruro de Litio , Ratones Noqueados , Poliuria/genética
9.
Kidney Int ; 87(6): 1164-75, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25629553

RESUMEN

Polycystic kidney diseases (PKDs) are inherited disorders characterized by the formation of fluid filled renal cysts. Elevated cAMP levels in PKDs stimulate progressive cyst enlargement involving cell proliferation and transepithelial fluid secretion often leading to end-stage renal disease. The glycogen synthase kinase-3 (GSK3) family of protein kinases consists of GSK3α and GSK3ß isoforms and has a crucial role in multiple cellular signaling pathways. We previously found that GSK3ß, a regulator of cell proliferation, is also crucial for cAMP generation and vasopressin-mediated urine concentration by the kidneys. However, the role of GSK3ß in the pathogenesis of PKDs is not known. Here we found that GSK3ß expression and activity were markedly upregulated and associated with cyst-lining epithelia in the kidneys of mice and humans with PKD. Renal collecting duct-specific gene knockout of GSK3ß or pharmacological inhibition of GSK3 effectively slowed down the progression of PKD in mouse models of autosomal recessive or autosomal dominant PKD. GSK3 inactivation inhibited cAMP generation and cell proliferation resulting in reduced cyst expansion, improved renal function, and extended life span. GSK3ß inhibition also reduced pERK, c-Myc, and cyclin-D1, known mitogens in proliferation of cystic epithelial cells. Thus, GSK3ß has a novel functional role in PKD pathophysiology, and its inhibition may be therapeutically useful to slow down cyst expansion and progression of PKD.


Asunto(s)
AMP Cíclico/metabolismo , Quistes/metabolismo , Quistes/patología , Glucógeno Sintasa Quinasa 3/metabolismo , Enfermedades Renales Poliquísticas/enzimología , Animales , Proliferación Celular/efectos de los fármacos , Ciclina D1/metabolismo , Inhibidores Enzimáticos/farmacología , Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Humanos , Riñón/enzimología , Túbulos Renales Colectores/enzimología , Ratones , Ratones Noqueados , Tamaño de los Órganos/efectos de los fármacos , Enfermedades Renales Poliquísticas/patología , Enfermedades Renales Poliquísticas/fisiopatología , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/metabolismo , Tiadiazoles/farmacología
10.
J Biol Chem ; 288(7): 5223-31, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23283969

RESUMEN

The epithelial sodium channel (ENaC) participates in the regulation of plasma sodium and volume, and gain of function mutations in the human channel cause salt-sensitive hypertension. Roles for the arachidonic acid epoxygenase metabolites, the epoxyeicosatrienoic acids (EETs), in ENaC activity have been identified; however, their mechanisms of action remain unknown. In polarized M1 cells, 14,15-EET inhibited amiloride-sensitive apical to basolateral sodium transport as effectively as epidermal growth factor (EGF). The EET effects were associated with increased threonine phosphorylation of the ENaC ß and γ subunits and abolished by inhibitors of (a) mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/extracellular signal regulated kinases 1 and 2 (MEK/ERK1/2) and (b) EGF receptor signaling. CYP2C44 epoxygenase knockdown blunted the sodium transport effects of EGF, and its 14,15-EET metabolite rescued the knockdown phenotype. The relevance of these findings is indicated by (a) the hypertension that results in mice administered cetuximab, an inhibitor of EGF receptor binding, and (b) immunological data showing an association between the pressure effects of cetuximab and reductions in ENaCγ phosphorylation. These studies (a) identify an ERK1/2-dependent mechanism for ENaC inhibition by 14,15-EET, (b) point to ENaC as a proximal target for EET-activated ERK1/2 mitogenic kinases, (c) characterize a mechanistic commonality between EGF and epoxygenase metabolites as ENaC inhibitors, and (d) suggest a CYP2C epoxygenase-mediated pathway for the regulation of distal sodium transport.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Canales Epiteliales de Sodio/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica , Ácido 8,11,14-Eicosatrienoico/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Antihipertensivos/farmacología , Cetuximab , Sistema Enzimático del Citocromo P-450/metabolismo , Familia 2 del Citocromo P450 , Factor de Crecimiento Epidérmico/metabolismo , Humanos , Hipertensión , Riñón/metabolismo , Masculino , Ratones , Modelos Biológicos , Fosforilación
11.
Eur J Cell Biol ; 102(2): 151316, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37084657

RESUMEN

The expression of the angiotensin-converting enzyme 2 (ACE2) is altered in multiple chronic kidney diseases like hypertension and renal fibrosis, where the signaling from the basal membrane proteins is critical for the development and progression of the various pathologies. Integrins are heterodimeric cell surface receptors that have important roles in the progression of these chronic kidney diseases by altering various cell signaling pathways in response to changes in the basement membrane proteins. It is unclear whether integrin or integrin-mediated signaling affects the ACE2 expression in the kidney. The current study tests the hypothesis that integrin ß1 regulates the expression of ACE2 in kidney epithelial cells. The role of integrin ß1 in ACE2 expression in renal epithelial cells was investigated by shRNA-mediated knockdown and pharmacological inhibition. In vivo studies were carried out using epithelial cell-specific deletion of integrin ß1 in the kidneys. Deletion of integrin ß1 from the mouse renal epithelial cells reduced the expression of ACE2 in the kidney. Furthermore, the downregulation of integrin ß1 using shRNA decreased ACE2 expression in human renal epithelial cells. ACE2 expression levels were also decreased in renal epithelial cells and cancer cells when treated with an integrin α2ß1 antagonist, BTT 3033. SARS-CoV-2 viral entry to human renal epithelial cells and cancer cells was also inhibited by BTT 3033. This study demonstrates that integrin ß1 positively regulates the expression of ACE2, which is required for the entry of SARS-CoV-2 into kidney cells.


Asunto(s)
COVID-19 , Insuficiencia Renal Crónica , Humanos , Animales , Ratones , Integrina beta1/genética , Integrina beta1/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , SARS-CoV-2/metabolismo , COVID-19/metabolismo , COVID-19/patología , Riñón/metabolismo , Riñón/patología , Células Epiteliales/metabolismo , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología
12.
Kidney Int ; 82(9): 1000-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22785175

RESUMEN

Renal proximal tubular damage and repair are hallmarks of acute kidney injury. As glycogen synthase kinase-3ß (GSK3ß) is an important cellular regulator of survival and proliferation, we determined its role during injury and recovery of proximal tubules in a mercuric chloride-induced nephrotoxic model of acute kidney injury. Renal proximal tubule-specific GSK3ß knockout mice exposed to mercuric chloride had improved survival and renal function compared to wild-type mice. Apoptosis, measured by TUNEL staining, Bax activation, and caspase 3 cleavage, was reduced in the knockout mice. The restoration of renal structure, function, and cell proliferation was also accelerated in the GSK3ß knockout mice. This enhanced repair, evidenced by increased Ki-67 and BRDU staining, along with increased cyclin D1 and c-myc levels, was recapitulated by treatment of wild-type mice with the small-molecule GSK3 inhibitor TDZD-8 following injury. This confirmed that hastened repair in the knockout mice was not merely due to lower initial injury levels. Thus, inhibition of GSK3ß prior to nephrotoxic insult protects from renal injury. Such treatment after acute kidney injury may accelerate repair and regeneration.


Asunto(s)
Lesión Renal Aguda/fisiopatología , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Túbulos Renales Proximales/fisiología , Enfermedad Aguda , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/mortalidad , Lesión Renal Aguda/patología , Animales , Antiinfecciosos Locales/toxicidad , Apoptosis/fisiología , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Estimación de Kaplan-Meier , Túbulos Renales Proximales/patología , Cloruro de Mercurio/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tiadiazoles/farmacología
13.
Curr Opin Nephrol Hypertens ; 21(5): 541-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22691876

RESUMEN

PURPOSE OF REVIEW: Glycogen synthase kinase-3 (GSK3) is an enzyme that is gaining prominence as a critical signaling molecule in the epithelial cells of renal tubules. This review will focus on recent findings exploring the role of GSK3 in renal collecting ducts, especially its role in urine concentration involving vasopressin signaling. RECENT FINDINGS: Recent studies using inhibition or tissue-specific gene deletion of GSK3 revealed the mechanism by which GSK3 regulates aquaporin 2 water channels via adenylate cyclase or the prostaglandin-E2 pathway. In other studies, postnatal treatment with lithium, an inhibitor of GSK3, increased cell proliferation and led to microcyst formation in rat kidneys. These studies suggest that loss of GSK3 activity could interfere with renal water transport at two levels. In the short term, it could disrupt vasopressin signaling in collecting duct cells and in the long term it could alter the structure of the collecting ducts, making them less responsive to the hydro-osmotic effects of vasopressin. SUMMARY: Ongoing studies reveal the crucial role played by GSK3 in the regulation of vasopressin action in the renal collecting ducts and suggest a possible use of GSK3 inhibitors in disease conditions associated with disrupted vasopressin signaling.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Túbulos Renales Colectores/enzimología , Animales , Acuaporina 2/metabolismo , Transporte Biológico , Humanos , Transducción de Señal , Vasopresinas/metabolismo
14.
Nat Med ; 11(8): 861-6, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16007095

RESUMEN

Thiazolidinediones (TZDs) are widely used to treat type 2 diabetes mellitus; however, their use is complicated by systemic fluid retention. Along the nephron, the pharmacological target of TZDs, peroxisome proliferator-activated receptor-gamma (PPARgamma, encoded by Pparg), is most abundant in the collecting duct. Here we show that mice treated with TZDs experience early weight gain from increased total body water. Weight gain was blocked by the collecting duct-specific diuretic amiloride and was also prevented by deletion of Pparg from the collecting duct, using Pparg (flox/flox) mice. Deletion of collecting duct Pparg decreased renal Na(+) avidity and increased plasma aldosterone. Treating cultured collecting ducts with TZDs increased amiloride-sensitive Na(+) absorption and Scnn1g mRNA (encoding the epithelial Na(+) channel ENaCgamma) expression through a PPARgamma-dependent pathway. These studies identify Scnn1g as a PPARgamma target gene in the collecting duct. Activation of this pathway mediates fluid retention associated with TZDs, and suggests amiloride might provide a specific therapy.


Asunto(s)
Amilorida/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Nefronas/fisiología , Tiazolidinedionas/metabolismo , Tiazolidinedionas/farmacología , Equilibrio Hidroelectrolítico/fisiología , Análisis de Varianza , Animales , Análisis Químico de la Sangre , Líquidos Corporales/efectos de los fármacos , Peso Corporal , Inmunoprecipitación de Cromatina , Canales Epiteliales de Sodio , Marcación de Gen , Ratones , Ratones Transgénicos , Nefronas/efectos de los fármacos , PPAR gamma/genética , PPAR gamma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sodio/metabolismo , Canales de Sodio/genética , Canales de Sodio/metabolismo , Tiazolidinedionas/efectos adversos , Equilibrio Hidroelectrolítico/efectos de los fármacos
15.
J Am Soc Nephrol ; 21(3): 428-37, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20056751

RESUMEN

Glycogen synthase kinase 3beta (GSK3beta), a serine/threonine protein kinase, is a key target of drug discovery in several diseases, including diabetes and Alzheimer disease. Because lithium, a potent inhibitor of GSK3beta, causes nephrogenic diabetes insipidus, GSK3beta may play a crucial role in regulating water homeostasis. We developed renal collecting duct-specific GSK3beta knockout mice to determine whether deletion of GSK3beta affects arginine vasopressin-dependent renal water reabsorption. Although only mildly polyuric under normal conditions, knockout mice exhibited an impaired urinary concentrating ability in response to water deprivation or treatment with a vasopressin analogue. The knockout mice had reduced levels of mRNA, protein, and membrane localization of the vasopressin-responsive water channel aquaporin 2 compared with wild-type mice. The knockout mice also expressed lower levels of pS256-AQP2, a phosphorylated form crucial for membrane trafficking. Levels of cAMP, a major regulator of aquaporin 2 expression and trafficking, were also lower in the knockout mice. Both GSK3beta gene deletion and pharmacologic inhibition of GSK3beta reduced adenylate cyclase activity. In summary, GSK3beta inactivation or deletion reduces aquaporin 2 expression by modulating adenylate cyclase activity and cAMP generation, thereby impairing responses to vasopressin in the renal collecting duct.


Asunto(s)
Adenilil Ciclasas/metabolismo , Fármacos Antidiuréticos/farmacología , Desamino Arginina Vasopresina/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Capacidad de Concentración Renal/fisiología , Túbulos Renales Colectores/enzimología , Adenilil Ciclasas/genética , Animales , Acuaporina 2/genética , Acuaporina 2/metabolismo , Células Cultivadas , Colforsina/farmacología , AMP Cíclico/metabolismo , Femenino , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Capacidad de Concentración Renal/efectos de los fármacos , Túbulos Renales Colectores/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Privación de Agua/fisiología , Equilibrio Hidroelectrolítico/efectos de los fármacos , Equilibrio Hidroelectrolítico/fisiología
16.
Cell Death Dis ; 12(10): 947, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34650051

RESUMEN

Autosomal-dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease and is characterized by progressive growth of fluid-filled cysts. Growth factors binding to receptor tyrosine kinases (RTKs) stimulate cell proliferation and cyst growth in PKD. Nintedanib, a triple RTK inhibitor, targets the vascular endothelial growth-factor receptor (VEGFR), platelet-derived growth-factor receptor (PDGFR), and fibroblast growth-factor receptor (FGFR), and is an approved drug for the treatment of non-small-cell lung carcinoma and idiopathic lung fibrosis. To determine if RTK inhibition using nintedanib can slow ADPKD progression, we tested its effect on human ADPKD renal cyst epithelial cells and myofibroblasts in vitro, and on Pkd1f/fPkhd1Cre and Pkd1RC/RC, orthologous mouse models of ADPKD. Nintedanib significantly inhibited cell proliferation and in vitro cyst growth of human ADPKD renal cyst epithelial cells, and cell viability and migration of human ADPKD renal myofibroblasts. Consistently, nintedanib treatment significantly reduced kidney-to-body-weight ratio, renal cystic index, cystic epithelial cell proliferation, and blood-urea nitrogen levels in both the Pkd1f/fPkhd1Cre and Pkd1RC/RC mice. There was a corresponding reduction in ERK, AKT, STAT3, and mTOR activity and expression of proproliferative factors, including Yes-associated protein (YAP), c-Myc, and Cyclin D1. Nintedanib treatment significantly reduced fibrosis in Pkd1RC/RC mice, but did not affect renal fibrosis in Pkd1f/fPkhd1Cre mice. Overall, these results suggest that nintedanib may be repurposed to effectively slow cyst growth in ADPKD.


Asunto(s)
Indoles/uso terapéutico , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Indoles/farmacología , Riñón/efectos de los fármacos , Riñón/patología , Ratones Noqueados , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Superficie Celular/metabolismo , Transducción de Señal/efectos de los fármacos
17.
Nephron ; 144(12): 609-612, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32726778

RESUMEN

Acute kidney injury (AKI) is a common clinical syndrome that involves renal tubular epithelial cell death and leads to acute decline in renal function. Improper tubular regeneration following AKI often leads to CKD. We discuss the role of a serine/threonine protein kinase called glycogen synthase kinase-3 (GSK3) in renal tubular injury and renal fibrosis. We also highlight the importance of GSK3 as a potential drug target in AKI patients and molecular mechanisms promoting tissue regeneration.


Asunto(s)
Lesión Renal Aguda/enzimología , Glucógeno Sintasa Quinasa 3/metabolismo , Transducción de Señal , Animales , Apoptosis , Células Epiteliales/patología , Humanos , Túbulos Renales/patología
18.
Oncogene ; 39(6): 1231-1245, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31616061

RESUMEN

Arginine vasopressin (AVP) and its type-2 receptor (V2R) play an essential role in the regulation of salt and water homeostasis by the kidneys. V2R activation also stimulates proliferation of renal cell carcinoma (RCC) cell lines in vitro. The current studies investigated V2R expression and activity in human RCC tumors, and its role in RCC tumor growth. Examination of the cancer genome atlas (TCGA) database, and analysis of human RCC tumor tissue microarrays, cDNA arrays and tumor biopsy samples demonstrated V2R expression and activity in clear cell RCC (ccRCC). In vitro, V2R antagonists OPC31260 and Tolvaptan, or V2R gene silencing reduced wound closure and cell viability of 786-O and Caki-1 human ccRCC cell lines. Similarly in mouse xenograft models, Tolvaptan and OPC31260 decreased RCC tumor growth by reducing cell proliferation and angiogenesis, while increasing apoptosis. In contrast, the V2R agonist dDAVP significantly increased tumor growth. High intracellular cAMP levels and ERK1/2 activation were observed in human ccRCC tumors. In mouse tumors and Caki-1 cells, V2R agonists reduced cAMP and ERK1/2 activation, while dDAVP treatment had the reverse effect. V2R gene silencing in Caki-1 cells also reduced cAMP and ERK1/2 activation. These results provide novel evidence for a pathogenic role of V2R signaling in ccRCC, and suggest that inhibitors of the AVP-V2R pathway, including the FDA-approved drug Tolvaptan, could be utilized as novel ccRCC therapeutics.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Renales/tratamiento farmacológico , Receptores de Vasopresinas/química , Tolvaptán/farmacología , Animales , Antagonistas de los Receptores de Hormonas Antidiuréticas/farmacología , Apoptosis , Biomarcadores de Tumor , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Estudios de Casos y Controles , Ciclo Celular , Proliferación Celular , Femenino , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Ratones , Ratones Desnudos , Pronóstico , Receptores de Vasopresinas/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Mol Cell Biol ; 8(6): 464-476, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27190311

RESUMEN

Glycogen synthase kinase 3ß (GSK3ß), a serine/threonine protein kinase, is commonly known to be regulated at the level of its activity. However, in some diseases including polycystic kidney disease (PKD), GSK3ß expression is increased and plays a pathophysiological role. The current studies aimed to determine the mechanism for the increased GSK3ß expression in PKD and its significance to disease progression. In mouse models of PKD, increases in renal GSK3ß corresponded with increases in renal cAMP levels and disease progression. In vivo and in vitro studies revealed that GSK3ß is a cAMP-responsive gene, and elevated cAMP levels, as seen in PKD, can increase GSK3ß expression. In normal mice, vasopressin signaling induced by water deprivation increased GSK3ß expression, which decreased following rehydration. Examination of the GSK3ß promoter revealed five potential binding sites for the transcription factor, cAMP response element binding protein (CREB). CREB was found to bind to GSK3ß promoter and essential for cAMP-mediated regulation of GSK3ß. Importantly, this regulation was demonstrated to be part of a feed-forward loop in which cAMP through CREB regulates GSK3ß expression, and GSK3ß in turn positively regulates cAMP generation. GSK3ß or CREB inhibition reduced transepithelial fluid secretion and cyst expansion in vitro Thus, disruption at any point of this destructive cycle may be therapeutically useful to reduce cyst expansion and preserve renal function in PKD.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Enfermedades Renales Poliquísticas/metabolismo , Animales , Líquidos Corporales/metabolismo , AMP Cíclico , Perros , Técnicas de Inactivación de Genes , Glucógeno Sintasa Quinasa 3 beta/genética , Humanos , Riñón/enzimología , Riñón/patología , Células de Riñón Canino Madin Darby , Ratones Endogámicos C57BL , Enfermedades Renales Poliquísticas/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Canales Catiónicos TRPP , Vasopresinas/metabolismo
20.
Biochim Biophys Acta ; 1634(1-2): 15-23, 2003 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-14563409

RESUMEN

Prostaglandin E(2) (PGE(2)) plays an important role in genitourinary function. Multiple enzymes are involved in its biosynthesis. Here we report the genomic structure and tissue-selective expression of cytosolic PGE(2) synthase (cPGES) in genitourinary tissues. Full-length mouse cPGES cDNA was cloned by reverse transcript-polymerase chain reaction (RT-PCR) and 5'- and 3'-rapid amplification of cDNA ends (RACE). Analysis of a cPGES cDNA with partially sequenced cPGES genomic clones and bioinformatic databases demonstrates that the murine cPGES gene spans approximately 22 kb and consists of eight exons. The cPGES gene promoter is GC-rich and contains many SP1 sites but lacks an obvious TATA box motif. RNase protection assay revealed constitutive expression of cPGES was greatest in the testis with lower levels in the ovary, kidney, bladder and uterus. In situ hybridization studies demonstrated that cPGES mRNA was most highly expressed in the epithelial cells of seminiferous tubules in the testis. In the female reproductive tissues, cPGES was mainly localized in ovarian primary and secondary follicles and oviductal epithelial cells with less expression in uterine endometrium. In the kidney cPGES expression was diffusely expressed. In urinary bladder, cPGES expression was restricted to the transitional epithelial cells. This expression pattern is consistent with an important role for cPGES-mediated PGE(2) in urogenital tissue function.


Asunto(s)
Prostaglandina-Endoperóxido Sintasas/genética , Prostaglandina-Endoperóxido Sintasas/metabolismo , Sistema Urogenital/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Clonación Molecular , Femenino , Hibridación in Situ , Ratones , Datos de Secuencia Molecular , ARN Mensajero/metabolismo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA