Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-38260358

RESUMEN

Polycystin-1 (PC1) is the membrane protein product of the PKD1 gene whose mutation is responsible for 85% of the cases of autosomal dominant polycystic kidney disease (ADPKD). ADPKD is primarily characterized by the formation of renal cysts and potential kidney failure. PC1 is an atypical G protein-coupled receptor (GPCR) consisting of 11 transmembrane helices and an autocatalytic GAIN domain that cleaves PC1 into extracellular N-terminal (NTF) and membrane-embedded C-terminal (CTF) fragments. Recently, signaling activation of the PC1 CTF was shown to be regulated by a stalk tethered agonist (TA), a distinct mechanism observed in the adhesion GPCR family. A novel allosteric activation pathway was elucidated for the PC1 CTF through a combination of Gaussian accelerated molecular dynamics (GaMD), mutagenesis and cellular signaling experiments. Here, we show that synthetic, soluble peptides with 7 to 21 residues derived from the stalk TA, in particular, peptides including the first 9 residues (p9), 17 residues (p17) and 21 residues (p21) exhibited the ability to re-activate signaling by a stalkless PC1 CTF mutant in cellular assays. To reveal molecular mechanisms of stalk peptide-mediated signaling activation, we have applied a novel Peptide GaMD (Pep-GaMD) algorithm to elucidate binding conformations of selected stalk peptide agonists p9, p17 and p21 to the stalkless PC1 CTF. The simulations revealed multiple specific binding regions of the stalk peptide agonists to the PC1 protein including an "intermediate" bound yet inactive state. Our Pep-GaMD simulation findings were consistent with the cellular assay experimental data. Binding of peptide agonists to the TOP domain of PC1 induced close TOP-putative pore loop interactions, a characteristic feature of the PC1 CTF signaling activation mechanism. Using sequence covariation analysis of PC1 homologs, we further showed that the peptide binding regions were consistent with covarying residue pairs identified between the TOP domain and the stalk TA. Therefore, structural dynamic insights into the mechanisms of PC1 activation by stalk-derived peptide agonists have enabled an in-depth understanding of PC1 signaling. They will form a foundation for development of PC1 as a therapeutic target for the treatment of ADPKD.

2.
Am J Physiol Renal Physiol ; 323(4): F492-F506, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35979967

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is a debilitating renal neoplastic disorder with limited treatment options. It is characterized by the formation of large fluid-filled cysts that develop from kidney tubules through abnormal cell proliferation and cyst-filling fluid secretion driven by cAMP-dependent Cl- secretion. We tested the effectiveness of the indazole carboxylic acid H2-gamendazole (H2-GMZ), a derivative of lonidamine, to inhibit these processes using in vitro and in vivo models of ADPKD. H2-GMZ was effective in rapidly blocking forskolin-induced, Cl--mediated short-circuit currents in human ADPKD cells, and it significantly inhibited both cAMP- and epidermal growth factor-induced proliferation of ADPKD cells. Western blot analysis of H2-GMZ-treated ADPKD cells showed decreased phosphorylated ERK and decreased hyperphosphorylated retinoblastoma levels. H2-GMZ treatment also decreased ErbB2, Akt, and cyclin-dependent kinase 4, consistent with inhibition of heat shock protein 90, and it decreased levels of the cystic fibrosis transmembrane conductance regulator Cl- channel protein. H2-GMZ-treated ADPKD cultures contained a higher proportion of smaller cells with fewer and smaller lamellipodia and decreased cytoplasmic actin staining, and they were unable to accomplish wound closure even at low H2-GMZ concentrations, consistent with an alteration in the actin cytoskeleton and decreased cell motility. Experiments using mouse metanephric organ cultures showed that H2-GMZ inhibited cAMP-stimulated cyst growth and enlargement. In vivo, H2-GMZ was effective in slowing postnatal cyst formation and kidney enlargement in the Pkd1flox/flox: Pkhd1-Cre mouse model. Thus, H2-GMZ treatment decreases Cl- secretion, cell proliferation, cell motility, and cyst growth. These properties, along with its reported low toxicity, suggest that H2-GMZ might be an attractive candidate for treatment of ADPKD.NEW & NOTEWORTHY Autosomal dominant polycystic kidney disease (ADPKD) is a renal neoplastic disorder characterized by the formation of large fluid-filled cysts that develop from kidney tubules through abnormal cell proliferation and cyst-filling fluid secretion driven by cAMP-dependent Cl- secretion. This study shows that the lonidamine derivative H2-GMZ inhibits Cl- secretion, cell proliferation, and cyst growth, suggesting that it might have therapeutic value for the treatment of ADPKD.


Asunto(s)
Quistes , Enfermedades Renales Poliquísticas , Riñón Poliquístico Autosómico Dominante , Actinas/metabolismo , Animales , Ácidos Carboxílicos/metabolismo , Proliferación Celular , Células Cultivadas , Colforsina/farmacología , Quinasa 4 Dependiente de la Ciclina/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Quistes/metabolismo , Familia de Proteínas EGF/metabolismo , Proteínas de Choque Térmico/metabolismo , Humanos , Indazoles/metabolismo , Indazoles/farmacología , Riñón/metabolismo , Ratones , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Enfermedades Renales Poliquísticas/metabolismo , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Superficie Celular
3.
Proc Natl Acad Sci U S A ; 119(19): e2113786119, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35522707

RESUMEN

Polycystin-1 (PC1) is an important unusual G protein-coupled receptor (GPCR) with 11 transmembrane domains, and its mutations account for 85% of cases of autosomal dominant polycystic kidney disease (ADPKD). PC1 shares multiple characteristics with Adhesion GPCRs. These include a GPCR proteolysis site that autocatalytically divides these proteins into extracellular, N-terminal, and membrane-embedded, C-terminal fragments (CTF), and a tethered agonist (TA) within the N-terminal stalk of the CTF that is suggested to activate signaling. However, the mechanism by which a TA can activate PC1 is not known. Here, we have combined functional cellular signaling experiments of PC1 CTF expression constructs encoding wild type, stalkless, and three different ADPKD stalk variants with all-atom Gaussian accelerated molecular dynamics (GaMD) simulations to investigate TA-mediated signaling activation. Correlations of residue motions and free-energy profiles calculated from the GaMD simulations correlated with the differential signaling abilities of wild type and stalk variants of PC1 CTF. They suggested an allosteric mechanism involving residue interactions connecting the stalk, Tetragonal Opening for Polycystins (TOP) domain, and putative pore loop in TA-mediated activation of PC1 CTF. Key interacting residues such as N3074­S3585 and R3848­E4078 predicted from the GaMD simulations were validated by mutagenesis experiments. Together, these complementary analyses have provided insights into a TA-mediated activation mechanism of PC1 CTF signaling, which will be important for future rational drug design targeting PC1.


Asunto(s)
Riñón Poliquístico Autosómico Dominante , Canales Catiónicos TRPP , Femenino , Humanos , Masculino , Mutación , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Transducción de Señal , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
4.
Methods Cell Biol ; 153: 169-183, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31395378

RESUMEN

Metanephric organ culture, or ex vivo embryonic kidney culture, was developed in the mid-twentieth century as a means to understand the development of the mammalian kidney and was used in early studies of polycystic kidney disease to explore mechanisms of renal cyst initiation by non-genetic factors. Following the identification of cystogenic genes, a resurgence of the use of metanephric organ culture occurred and has yielded insight into basic mechanisms of cystic dilation; facilitated identification of pathogenic pathways and potential therapeutic targets; and provided a system for evaluating therapeutic agents. This chapter provides detailed, step-by-step protocols with rationale and tips for the establishment, maintenance and treatment of metanephric organ cultures, and for performance of the most commonly employed secondary analyses of these cultures.


Asunto(s)
Embrión de Mamíferos , Riñón , Técnicas de Cultivo de Órganos/métodos , Enfermedades Renales Poliquísticas/patología , Animales , Medios de Cultivo/metabolismo , AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Microscopía Intravital/instrumentación , Microscopía Intravital/métodos , Ratones , Microdisección/instrumentación , Microdisección/métodos , Microscopía Fluorescente/instrumentación , Microscopía Fluorescente/métodos
5.
Hum Mol Genet ; 27(19): 3313-3324, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29931260

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the growth of renal cysts that ultimately destroy kidney function. Mutations in the PKD1 and PKD2 genes cause ADPKD. Their protein products, polycystin-1 (PC1) and polycystin-2 (PC2) have been proposed to form a calcium-permeable receptor-channel complex; however the mechanisms by which they function are almost completely unknown. Most mutations in PKD1 are truncating loss-of-function mutations or affect protein biogenesis, trafficking or stability and reveal very little about the intrinsic biochemical properties or cellular functions of PC1. An ADPKD patient mutation (L4132Δ or ΔL), resulting in a single amino acid deletion in a putative G-protein binding region of the PC1 C-terminal cytosolic tail, was found to significantly decrease PC1-stimulated, G-protein-dependent signaling in transient transfection assays. Pkd1ΔL/ΔL mice were embryo-lethal suggesting that ΔL is a functionally null mutation. Kidney-specific Pkd1ΔL/cond mice were born but developed severe, postnatal cystic disease. PC1ΔL protein expression levels and maturation were comparable to those of wild type PC1, and PC1ΔL protein showed cell surface localization. Expression of PC1ΔL and PC2 complexes in transfected CHO cells failed to support PC2 channel activity, suggesting that the role of PC1 is to activate G-protein signaling to regulate the PC1/PC2 calcium channel.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas/genética , Riñón Poliquístico Autosómico Dominante/genética , Canales Catiónicos TRPP/genética , Animales , Células CHO , Canales de Calcio/genética , Cilios/genética , Cilios/patología , Cricetulus , Humanos , Riñón/patología , Ratones , Mutación , Riñón Poliquístico Autosómico Dominante/patología , Dominios Proteicos/genética , Transducción de Señal
6.
J Am Soc Nephrol ; 27(4): 1124-34, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26289612

RESUMEN

cAMP stimulates cell proliferation and Cl(-)-dependent fluid secretion, promoting the progressive enlargement of renal cysts in autosomal dominant polycystic kidney disease (ADPKD). Intracellular cAMP levels are determined by the balance of cAMP synthesis by adenylyl cyclases and degradation by phosphodiesterases (PDEs). Therefore, PDE isoform expression and activity strongly influence global and compartmentalized cAMP levels. We report here that PDE3 and PDE4 expression levels are lower in human ADPKD tissue and cells compared with those of normal human kidneys (NHKs), whereas PDE1 levels are not significantly different. Inhibition of PDE4 caused a greater increase in basal and vasopressin (AVP)-stimulated cAMP levels and Cl(-) secretion by ADPKD cells than inhibition of PDE1, and inhibition of PDE4 induced cyst-like dilations in cultured mouse Pkd1(-/-) embryonic kidneys. In contrast, inhibition of PDE1 caused greater stimulation of extracellular signal-regulated kinase (ERK) and proliferation of ADPKD cells than inhibition of PDE4, and inhibition of PDE1 enhanced AVP-induced ERK activation. Notably, inhibition of PDE1, the only family of Ca(2+)-regulated PDEs, also induced a mitogenic response to AVP in NHK cells, similar to the effect of restricting intracellular Ca(2+). PDE1 coimmunoprecipitated with B-Raf and A-kinase anchoring protein 79, and AVP increased this interaction in ADPKD but not NHK cells. These data suggest that whereas PDE4 is the major PDE isoform involved in the regulation of global intracellular cAMP and Cl(-) secretion, PDE1 specifically affects the cAMP signal to the B-Raf/MEK/ERK pathway and regulates AVP-induced proliferation of ADPKD cells.


Asunto(s)
Proliferación Celular/fisiología , Líquido Extracelular/metabolismo , Hidrolasas Diéster Fosfóricas/fisiología , Riñón Poliquístico Autosómico Dominante/patología , Riñón Poliquístico Autosómico Dominante/fisiopatología , Animales , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Isoenzimas/fisiología , Ratones
7.
J Membr Biol ; 248(6): 1145-57, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26289599

RESUMEN

Cyst enlargement in autosomal dominant polycystic kidney disease (ADPKD) requires the transepithelial secretion of fluid into the cyst lumen. We previously showed that physiological amounts of ouabain enhance cAMP-dependent fluid secretion and cyst growth of human ADPKD cyst epithelial cells in culture and formation of cyst-like dilations in metanephric kidneys from Pkd1 mutant mice. Here, we investigated the mechanisms by which ouabain promotes cAMP-dependent fluid secretion and cystogenesis. Ouabain (3 nM) enhanced cAMP-induced cyst-like dilations in embryonic kidneys from Pkd1 (m1Bei) mice, but had no effect on metanephroi from Pkd1 (m1Bei) mice that lack expression of the cystic fibrosis transmembrane conductance regulator (CFTR). Similarly, ouabain stimulation of cAMP-induced fluid secretion and in vitro cyst growth of ADPKD cells were abrogated by CFTR inhibition, showing that CFTR is required for ouabain effects on ADPKD fluid secretion. Moreover, ouabain directly enhanced the cAMP-dependent Cl(-) efflux mediated by CFTR in ADPKD monolayers. Ouabain increased the trafficking of CFTR to the plasma membrane and up-regulated the expression of the CFTR activator PDZK1. Finally, ouabain decreased plasma membrane expression and activity of the Na,K-ATPase in ADPKD cells. Altogether, these results show that ouabain enhances net fluid secretion and cyst formation by activating apical anion secretion via CFTR and decreasing basolateral Na(+) transport via Na,K-ATPase. These results provide new information on the mechanisms by which ouabain affects ADPKD cells and further highlight the importance of ouabain as a non-genomic stimulator of cystogenesis in ADPKD.


Asunto(s)
Aniones/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Ouabaína/farmacología , Riñón Poliquístico Autosómico Dominante/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Membrana Celular/metabolismo , Cloruros/metabolismo , Colforsina/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Riñón Poliquístico Autosómico Dominante/genética , Cultivo Primario de Células , Simportadores de Cloruro de Sodio-Potasio/metabolismo
8.
J Membr Biol ; 246(7): 581-90, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23784065

RESUMEN

Cells derived from renal cysts of patients with autosomal dominant polycystic kidney disease (ADPKD) are abnormally sensitive to ouabain, responding to physiological ouabain concentrations with enhanced proliferation and increased forskolin-induced transepithelial fluid secretion. This requires activation of the epidermal growth factor receptor (EGFR), Src kinase and the extracellular signal-regulated kinases MEK and ERK. Here, we have determined if the ADPKD phenotype obtained in mouse cortical collecting duct cells by stable overexpression of the C-terminal domain of polycystin-1 (PC-1 C-tail) also elicits the ADPKD-like response to ouabain in the cells. M-1 C20 cells expressing the PC-1 C-tail and M-1 C17 cells lacking expression of this construct were treated with physiological concentrations of ouabain, and cell proliferation, activation of the EGFR-Src-MEK-ERK pathway, forskolin-induced transepithelial Cl(-) secretion and the sensitivity of Na,K-ATPase to ouabain were explored. M-1 C20 cells responded to ouabain with increased cell proliferation and ERK phosphorylation. Ouabain also augmented forskolin-induced and cystic fibrosis transmembrane conductance regulator-mediated apical secretion of Cl(-) in M-1 C20 cells. These effects required activation of EGFR, Src and MEK. In contrast, ouabain had no significant effects on M-1 C17 cells. Interestingly, approximately 20% of the Na,K-ATPase from M-1 C20 cells presented an abnormally increased sensitivity to ouabain. Overexpression of PC-1 C-tail in M-1 C20 cells is associated with an ouabain-sensitive phenotype and an increased ability of the cells to proliferate and secrete anions upon ouabain stimulation. This phenotype mimics the ouabain sensitivity of ADPKD cells and may help promote their cystogenic potential.


Asunto(s)
Resistencia a Medicamentos/genética , Expresión Génica , Ouabaína/farmacología , Dominios y Motivos de Interacción de Proteínas/genética , Canales Catiónicos TRPP/genética , Animales , Aniones/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Colforsina/farmacología , Receptores ErbB/metabolismo , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Colectores/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Canales Catiónicos TRPP/química
9.
Am J Physiol Renal Physiol ; 303(7): F982-90, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22859406

RESUMEN

In autosomal-dominant polycystic kidney disease (ADPKD), renal cysts develop by aberrant epithelial cell proliferation and transepithelial fluid secretion. We previously showed that ouabain increases proliferation of cultured human ADPKD cells via stimulation of the EGF receptor (EGFR)-Src-MEK/ERK signaling pathway. We examined whether ouabain affects fluid secretion and in vitro cyst growth of human ADPKD cell monolayers, ADPKD cell microcysts cultured in a three-dimensional collagen matrix, and metanephric organ cultures from Pkd1(m1Bei) mice. Physiological concentrations of ouabain alone did not affect net transepithelial basal-to-apical fluid transport in ADPKD monolayers or growth of cultured ADPKD microcysts. In contrast, in the presence of forskolin or 8-bromo-cAMP, ouabain significantly enhanced ADPKD fluid secretion and microcyst expansion. Ouabain exerted this effect by enhancing cAMP-dependent Cl(-) secretion via the CFTR. Similarly, ouabain accelerated cAMP-dependent cyst enlargement in Pkd1(m1Bei) mice metanephroi, with a more prominent response in homozygous than heterozygous mice. Ouabain had no effect on fluid secretion and cystogenesis of normal human kidney cells and caused only slight cystic dilations in wild-type mouse kidneys. The effects of ouabain in ADPKD cells and Pkd1(m1Bei) metanephroi were prevented by inhibitors of EGFR (AG1478), Src (PP2), and MEK (U0126). Together, our results show that ouabain, used in physiological concentrations, has synergistic effects on cAMP-mediated fluid secretion and cyst growth, via activation of the EGFR-Src-MEK pathway. These data provide important evidence for the role of ouabain as an endogenous hormone that exacerbates ADPKD cyst progression.


Asunto(s)
AMP Cíclico/metabolismo , Quistes/metabolismo , Receptores ErbB/metabolismo , Riñón/metabolismo , Ouabaína/metabolismo , Riñón Poliquístico Autosómico Dominante/metabolismo , Transducción de Señal/fisiología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colforsina/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Riñón/citología , Riñón/efectos de los fármacos , Ratones , Transducción de Señal/efectos de los fármacos
10.
J Biol Chem ; 285(50): 38818-31, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-20923779

RESUMEN

Mitogen-activated protein kinase (MAPK) cascades regulate a wide variety of cellular processes that ultimately depend on changes in gene expression. We have found a novel mechanism whereby one of the key MAP3 kinases, Mekk1, regulates transcriptional activity through an interaction with p53. The tumor suppressor protein p53 down-regulates a number of genes, including the gene most frequently mutated in autosomal dominant polycystic kidney disease (PKD1). We have discovered that Mekk1 translocates to the nucleus and acts as a co-repressor with p53 to down-regulate PKD1 transcriptional activity. This repression does not require Mekk1 kinase activity, excluding the need for an Mekk1 phosphorylation cascade. However, this PKD1 repression can also be induced by the stress-pathway stimuli, including TNFα, suggesting that Mekk1 activation induces both JNK-dependent and JNK-independent pathways that target the PKD1 gene. An Mekk1-p53 interaction at the PKD1 promoter suggests a new mechanism by which abnormally elevated stress-pathway stimuli might directly down-regulate the PKD1 gene, possibly causing haploinsufficiency and cyst formation.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Regiones Promotoras Genéticas , Canales Catiónicos TRPP/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Secuencia de Bases , Células COS , Núcleo Celular/metabolismo , Chlorocebus aethiops , Humanos , Ratones , Microscopía Fluorescente/métodos , Datos de Secuencia Molecular , Mutagénesis , Estrés Oxidativo , Factor de Necrosis Tumoral alfa/metabolismo
11.
Am J Physiol Renal Physiol ; 295(6): F1845-54, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18922886

RESUMEN

The retinoic acids all-trans retinoic acid (AT-RA) and 9-cis retinoic acid (9C-RA) and the retinoic acid receptors RAR and RXR significantly induce transcriptional activity from a 200-bp PKD1 proximal promoter in transfected mammalian cells. This PKD1 promoter region contains Ets, p53, and GC box motifs, but lacks a canonical RAR/RXR motif. Mutagenesis of the Ets sites did not affect RA induction. In contrast, GC box mutations completely blocked stimulation by AT-RA and by RXRbeta or RARbeta. Mithramycin A, which prevents Sp1 binding, significantly reduced basal promoter activity and suppressed upregulation by AT-RA and RXR. The 200-bp proximal promoter could not be induced by AT-RA in Drosophila SL2 cells, which lack Sp1, but could be activated in these cells transfected with exogenous Sp1. Small interfering RNA knockdown of Sp1 in mammalian cells completely blocked RXRbeta upregulation of the promoter. These data indicate that induction of the PKD1 promoter by retinoic acid is mediated through Sp1 elements. RT-PCR showed that AT-RA treatment of HEK293T cells increased the levels of endogenous PKD1 RNA, and chromatin immunoprecipitation showed the presence of both RXR and Sp1 at the PKD1 proximal promoter. These results suggest that retinoids and their receptors may play a role in PKD1 gene regulation.


Asunto(s)
Regiones Promotoras Genéticas , Canales Catiónicos TRPP/genética , Activación Transcripcional/efectos de los fármacos , Tretinoina/farmacología , Tretinoina/fisiología , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Genes Reporteros , Humanos , Riñón/embriología , Luciferasas/genética , Datos de Secuencia Molecular , Plásmidos , Receptores de Ácido Retinoico/fisiología , Receptores X Retinoide/fisiología
12.
Nat Med ; 14(8): 863-8, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18552856

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is caused by heterozygous mutations in either PKD1 or PKD2, genes that encode polycystin-1 and polycystin-2, respectively. We show here that tumor necrosis factor-alpha (TNF-alpha), an inflammatory cytokine present in the cystic fluid of humans with ADPKD, disrupts the localization of polycystin-2 to the plasma membrane and primary cilia through a scaffold protein, FIP2, which is induced by TNF-alpha. Treatment of mouse embryonic kidney organ cultures with TNF-alpha resulted in formation of cysts, and this effect was exacerbated in the Pkd2(+/-) kidneys. TNF-alpha also stimulated cyst formation in vivo in Pkd2(+/-) mice. In contrast, treatment of Pkd2(+/-) mice with the TNF-alpha inhibitor etanercept prevented cyst formation. These data reveal a pathway connecting TNF-alpha signaling, polycystins and cystogenesis, the activation of which may reduce functional polycystin-2 below a critical threshold, precipitating the ADPKD cellular phenotype.


Asunto(s)
Proteínas del Ojo/genética , Mutación , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/inmunología , Canales Catiónicos TRPP/metabolismo , Factor de Transcripción TFIIIA/genética , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Proteínas de Ciclo Celular , Membrana Celular/metabolismo , Citocinas/metabolismo , Proteínas del Ojo/metabolismo , Genes Dominantes , Humanos , Inflamación , Riñón/metabolismo , Proteínas de Transporte de Membrana , Ratones , Técnicas de Cultivo de Órganos/métodos , Factor de Transcripción TFIIIA/metabolismo
13.
J Biol Chem ; 283(18): 12624-34, 2008 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-18321855

RESUMEN

Polycystin-1 (PC1) may play an important role in skeletogenesis through regulation of the bone-specific transcription factor Runx2-II. In the current study we found that PC1 co-localizes with the calcium channel polycystin-2 (PC2) in primary cilia of MC3T3-E1 osteoblasts. To establish the role of Runx2-II in mediating PC1 effects on bone, we crossed heterozygous Pkd1(m1Bei) and Runx2-II mice to create double heterozygous mice (Pkd1(+/m1Bei)/Runx2-II(+/-)) deficient in both PC1 and Runx2-II. Pkd1(+/m1Bei)/Runx2-II(+/-) mice exhibited additive reductions in Runx2-II expression that was associated with impaired endochondral bone development, defective osteoblast-mediated bone formation, and osteopenia. In addition, we found that basal intracellular calcium levels were reduced in homozygous Pkd1(m1Bei) osteoblasts. In contrast, overexpression of a PC1 C-tail construct increased intracellular calcium and selectively stimulated Runx2-II P1 promoter activity in osteoblasts through a calcium-dependent mechanism. Site-directed mutagenesis of critical amino acids in the coiled-coil domain of PC1 required for coupling to PC2 abolished PC1-mediated Runx2-II P1 promoter activity. Additional promoter analysis mapped the PC1-responsive region to the "osteoblast-specific" enhancer element between -420 and -350 bp that contains NFI and AP-1 binding sites. Chromatin immunoprecipitation assays confirmed the calcium-dependent binding of NFI to this region. These findings indicate that PC1 regulates osteoblast function through intracellular calcium-dependent control of Runx2-II expression. The overall function of the primary cilium-polycystin complex may be to sense and transduce environmental clues into signals regulating osteoblast differentiation and bone development.


Asunto(s)
Desarrollo Óseo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Osteoblastos/metabolismo , Canales Catiónicos TRPP/metabolismo , Factores de Transcripción/metabolismo , Animales , Animales Recién Nacidos , Secuencia de Bases , Huesos/anomalías , Huesos/embriología , Calcio/metabolismo , Línea Celular , Cilios/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Espacio Intracelular/metabolismo , Ratones , Datos de Secuencia Molecular , Tamaño de los Órganos , Especificidad de Órganos , Osteoblastos/patología , Regiones Promotoras Genéticas/genética , Transporte de Proteínas , Transducción de Señal , Factores de Transcripción/genética
14.
J Am Soc Nephrol ; 17(12): 3424-37, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17108316

RESUMEN

Metanephric organ culture has been used to determine whether embryonic kidney tubules can be stimulated by cAMP to form cysts. Under basal culture conditions, wild-type kidneys from embryonic day 13.5 to 15.5 mice grow in size and continue ureteric bud branching and tubule formation over a 4- to 5-d period. Treatment of these kidneys with 8-Br-cAMP or the cAMP agonist forskolin induced the formation of dilated tubules within 1 h, which enlarged over several days and resulted in dramatically expanded cyst-like structures of proximal tubule and collecting duct origin. Tubule dilation was reversible upon withdrawal of 8-Br-cAMP and was inhibited by the cAMP-dependent protein kinase inhibitor H89 and the cystic fibrosis transmembrane conductance regulator (CFTR) inhibitor CFTR(inh)172. For further testing of the role of CFTR, metanephric cultures were prepared from mice with a targeted mutation of the Cftr gene. In contrast to kidneys from wild-type mice, those from Cftr -/- mice showed no evidence of tubular dilation in response to 8-Br-cAMP, indicating that CFTR Cl(-) channels are functional in embryonic kidneys and are required for cAMP-driven tubule expansion. A requirement for transepithelial Cl(-) transport was demonstrated by inhibiting the basolateral Na(+),K(+),2Cl(-) co-transporter with bumetanide, which effectively blocked all cAMP-stimulated tubular dilation. For determination of whether cystic dilation occurs to a greater extent in PKD kidneys in response to cAMP, Pkd1(m1Bei) -/- embryonic kidneys were treated with 8-Br-cAMP and were found to form rapidly CFTR- and Na(+),K(+),2Cl(-) co-transporter-dependent cysts that were three- to six-fold larger than those of wild-type kidneys. These results suggest that cAMP can stimulate fluid secretion early in renal tubule development during the time when renal cysts first appear in PKD kidneys and that PKD-deficient renal tubules are predisposed to abnormally increased cyst expansion in response to elevated levels of cAMP.


Asunto(s)
8-Bromo Monofosfato de Adenosina Cíclica/farmacología , AMP Cíclico/fisiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Túbulos Renales/embriología , Enfermedades Renales Poliquísticas/embriología , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico , Amidas/farmacología , Animales , Benzoatos/farmacología , Colforsina/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/antagonistas & inhibidores , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/fisiopatología , Riñón Poliquístico Autosómico Dominante/etiología , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/fisiopatología , Simportadores de Cloruro de Sodio-Potasio/fisiología , Canales Catiónicos TRPP , Tiazolidinas/farmacología
15.
J Biol Chem ; 281(41): 30884-95, 2006 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-16905538

RESUMEN

We examined the osteoblast/osteocyte expression and function of polycystin-1 (PC1), a transmembrane protein that is a component of the polycystin-2 (PC2)-ciliary mechano-sensor complex in renal epithelial cells. We found that MC3T3-E1 osteoblasts and MLO-Y4 osteocytes express transcripts for PC1, PC2, and the ciliary proteins Tg737 and Kif3a. Immunohistochemical analysis detected cilia-like structures in MC3T3-E1 osteoblastic and MLO-Y4 osteocyte-like cell lines as well as primary osteocytes and osteoblasts from calvaria. Pkd1m1Bei mice have inactivating missense mutations of Pkd1 gene that encode PC1. Pkd1m1Bei homozygous mutant mice demonstrated delayed endochondral and intramembranous bone formation, whereas heterozygous Pkd1m1Bei mutant mice had osteopenia caused by reduced osteoblastic function. Heterozygous and homozygous Pkd1m1Bei mutant mice displayed a gene dose-dependent decrease in the expression of Runx2 and osteoblast-related genes. In addition, overexpression of constitutively active PC1 C-terminal constructs in MC3T3-E1 osteoblasts resulted in an increase in Runx2 P1 promoter activity and endogenous Runx2 expression as well as an increase in osteoblast differentiation markers. Conversely, osteoblasts derived from Pkd1m1Bei homozygous mutant mice had significant reductions in endogenous Runx2 expression, osteoblastic markers, and differentiation capacity ex vivo. Co-expression of constitutively active PC1 C-terminal construct into Pkd1m1Bei homozygous osteoblasts was sufficient to normalize Runx2 P1 promoter activity. These findings are consistent with a possible functional role of cilia and PC1 in anabolic signaling in osteoblasts/osteocytes.


Asunto(s)
Desarrollo Óseo , Cilios/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Osteoblastos/metabolismo , Osteocitos/metabolismo , Proteínas Quinasas/biosíntesis , Canales Catiónicos TRPP/química , Canales Catiónicos TRPP/metabolismo , Animales , Células Cultivadas , Ratones , Ratones Transgénicos , Mutación , Proteína Quinasa D2
16.
J Biol Chem ; 281(42): 31234-44, 2006 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-16931520

RESUMEN

This study provides evidence that the tumor suppressor protein, p53, is a transcriptional repressor of PKD1. Kidneys of p53-null mice expressed higher Pkd1 mRNA levels than wild-type littermates; gamma-irradiation suppressed PKD1 gene expression in p53+/+ but not p53-/- cells; and chromatin immunoprecipitation assays demonstrated the binding of p53 to the PKD1 promoter in vivo. In transient transfection assays, p53 repressed PKD1 promoter activity independently of endogenous p21. Deletion analysis mapped p53-mediated repression to the proximal promoter region of PKD1. Mutations of the DNA binding or C-terminal minimal repression domains of p53 abolished its ability to repress PKD1. Moreover, trichostatin A, an inhibitor of histone deacetylase activity, attenuated p53-induced repression of the PKD1 promoter. These findings, together with previous reports showing that dedifferentiated Pkd1-deficient cells express lower p53 and p21 levels, suggest a model whereby PKD1 signaling activates the p53-p21 differentiation pathway. In turn, p53 cooperates with histone deacetylases to repress PKD1 gene transcription. Loss of a p53-mediated negative feedback loop in PKD1 mutant cells may therefore contribute to deregulated PKD1 expression and cystogenesis.


Asunto(s)
Regulación de la Expresión Génica , Genes p53 , Canales Catiónicos TRPP/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/fisiología , Animales , Diferenciación Celular , Riñón/metabolismo , Ratones , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal
17.
Biochem Biophys Res Commun ; 342(4): 1005-13, 2006 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-16510125

RESUMEN

The Ets family of transcription factors consists of a group of highly conserved sequence-specific DNA binding proteins that functionally cooperate with other transcription factors to regulate a number of diverse cellular processes including proliferation, differentiation, and apoptosis. We have analyzed a 3.3kb 5'-upstream region of the human PKD1 promoter, using transient transfection in HEK293T cells and Drosophila SL2 cells, to demonstrate that the PKD1 promoter is a target of Ets family transcription factors. Our studies showed that PKD1 promoter-luciferase reporter gene expression is downregulated by cotransfected Fli-1 and is upregulated by cotransfected Ets-1. Using deletion constructs, we demonstrated that the sequences responding to Fli-1 and Ets-1 lie within the -200 to +33bp proximal promoter. This region was found to contain two putative Ets response elements (EREs): an upstream (Ets-A) sequence 5'-CGGAA-3' (-181 to -185) and a downstream (Ets-B) sequence 5'-CGGAT-3' (-129 to -133). Site-directed mutagenesis indicated that both EREs are functional. A Fli-1 DNA binding domain mutant construct (W321R), which is incapable of binding DNA, was unable to inhibit basal promoter activity. In contrast, a Fli-1 DNA binding domain truncation mutant construct, which only contains the DNA binding domain and lacks the transactivation domain, was able to inhibit. These results suggest that the effect of Fli-1 is through direct binding to these EREs. Direct binding of Fli-1 and Ets-1 to the Ets-A and Ets-B sites was supported by electrophoretic mobility shift assays. Lastly, competition between Fli-1 and Ets-1 for the two EREs was demonstrated by showing that increasing amounts of Ets-1 could overcome Fli-1 repression of promoter activity. Taken together, these experiments define the proximal PKD1 promoter region as a potential target of Ets family transcription factors.


Asunto(s)
Gelsolina/genética , Regiones Promotoras Genéticas/genética , Proteínas/genética , Proteínas Proto-Oncogénicas c-ets/genética , Receptores Citoplasmáticos y Nucleares/genética , Activación Transcripcional/genética , Animales , Células Cultivadas , Drosophila , Regulación de la Expresión Génica/genética , Proteínas de Microfilamentos , Canales Catiónicos TRPP , Transactivadores
18.
J Biol Chem ; 279(53): 55455-64, 2004 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-15466861

RESUMEN

Regulation of intracellular Ca(2+) mobilization has been associated with the functions of polycystin-1 (PC1) and polycystin-2 (PC2), the protein products of the PKD1 and PKD2 genes. We have now demonstrated that PC1 can activate the calcineurin/NFAT (nuclear factor of activated T-cells) signaling pathway through Galpha(q) -mediated activation of phospholipase C (PLC). Transient transfection of HEK293T cells with an NFAT promoter-luciferase reporter demonstrated that membrane-targeted PC1 constructs containing the membrane proximal region of the C-terminal tail, which includes the heterotrimeric G protein binding and activation domain, can stimulate NFAT luciferase activity. Inhibition of glycogen synthase kinase-3beta by LiCl treatment further increased PC1-mediated NFAT activity. PC1-mediated activation of NFAT was completely inhibited by the calcineurin inhibitor, cyclosporin A. Cotransfection of a construct expressing the Galpha(q) subunit augmented PC1-mediated NFAT activity, whereas the inhibitors of PLC (U73122) and the inositol trisphosphate and ryanodine receptors (xestospongin and 2-aminophenylborate) and a nonspecific Ca(2+) channel blocker (gadolinium) diminished PC1-mediated NFAT activity. PC2 was not able to activate NFAT. An NFAT-green fluorescent protein nuclear localization assay demonstrated that PC1 constructs containing the C-tail only or the entire 11-transmembrane spanning region plus C-tail induced NFAT-green fluorescent protein nuclear translocation. NFAT expression was demonstrated in the M-1 mouse cortical collecting duct cell line and in embryonic and adult mouse kidneys by reverse transcriptase-PCR and immunolocalization. These data suggest a model in which PC1 signaling leads to a sustained elevation of intracellular Ca(2+) mediated by PC1 activation of Galpha(q) followed by PLC activation, release of Ca(2+) from intracellular stores, and activation of store-operated Ca(2+) entry, thus activating calcineurin and NFAT.


Asunto(s)
Calcineurina/metabolismo , Proteínas/fisiología , Transducción de Señal , Transporte Activo de Núcleo Celular , Animales , Western Blotting , Ácidos Borónicos/farmacología , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio , Línea Celular , Núcleo Celular/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Estrenos/farmacología , Gadolinio/farmacología , Genes Reporteros , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Receptores de Inositol 1,4,5-Trifosfato , Riñón/embriología , Riñón/metabolismo , Cloruro de Litio/farmacología , Luciferasas/metabolismo , Compuestos Macrocíclicos , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Microscopía Fluorescente , Factores de Transcripción NFATC , Oxazoles/farmacología , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , Pirrolidinonas/farmacología , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Canales Catiónicos TRPP , Factores de Tiempo , Distribución Tisular , Transfección , Fosfolipasas de Tipo C/metabolismo
19.
J Biol Chem ; 279(39): 40419-30, 2004 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-15263001

RESUMEN

cAMP can be either mitogenic or anti-mitogenic, depending on the cell type. We demonstrated previously that cAMP inhibited the proliferation of normal renal epithelial cells and stimulated the proliferation of cells derived from the cysts of polycystic kidney disease (PKD) patients. The protein products of the genes causing PKD, polycystin-1 and polycystin-2, are thought to regulate intracellular calcium levels, suggesting that abnormal polycystin function may affect calcium signaling and thus cause a switch to the cAMP growth-stimulated phenotype. To test this hypothesis, we disrupted intracellular calcium mobilization by treating immortalized mouse M-1 collecting duct cells and primary cultures of human kidney epithelial cells with calcium channel blockers and by lowering extracellular calcium with EGTA. Calcium restriction for 3-5 h converted both cell types from a normal cAMP growth-inhibited phenotype to an abnormal cAMP growth-stimulated phenotype, characteristic of PKD. In M-1 cells, we showed that calcium restriction was associated with an elevation in B-Raf protein levels and cAMP-stimulated, Ras-dependent activation of B-Raf and ERK. Moreover, the activity of Akt, a negative regulator of B-Raf, was decreased by calcium restriction. Inhibition of Akt or phosphatidylinositol 3-kinase also allowed cAMP-dependent activation of B-Raf and ERK in normal calcium. These results suggest that calcium restriction causes an inhibition of the phosphatidylinositol 3-kinase/Akt pathway, which relieves the inhibition of B-Raf to allow the cAMP growth-stimulated phenotypic switch. Finally, M-1 cells stably overexpressing an inducible polycystin-1 C-terminal cytosolic tail construct were shown to exhibit a cAMP growth-stimulated phenotype involving B-Raf and ERK activation, which was reversed by the calcium ionophore A23187. We conclude that disruption of calcium mobilization in cells that are normally growth-inhibited by cAMP can derepress the B-Raf/ERK pathway, thus converting these cells to a phenotype that is growth-stimulated by cAMP.


Asunto(s)
Calcio/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Animales , Western Blotting , Calcimicina/farmacología , Bloqueadores de los Canales de Calcio/farmacología , División Celular , Línea Celular , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Ácido Egtácico/farmacología , Activación Enzimática , Células Epiteliales , Humanos , Ionóforos/farmacología , Riñón/citología , Riñón/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Modelos Biológicos , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Enfermedades Renales Poliquísticas/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogénicas B-raf , Canales Catiónicos TRPP , Factores de Tiempo , Transfección , Verapamilo/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA