Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Jan 06.
Article in English | MEDLINE | ID: mdl-38260358

ABSTRACT

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.
Front Mol Biosci ; 9: 1035507, 2022.
Article in English | MEDLINE | ID: mdl-36406261

ABSTRACT

Polycystin-1 (PC1) is an 11-transmembrane (TM) domain-containing protein encoded by the PKD1 gene, the most frequently mutated gene leading to autosomal dominant polycystic kidney disease (ADPKD). This large (> 462 kDal) protein has a complex posttranslational maturation process, with over five proteolytic cleavages having been described, and is found at multiple cellular locations. The initial description of the binding and activation of heterotrimeric Gαi/o by the juxtamembrane region of the PC1 cytosolic C-terminal tail (C-tail) more than 20 years ago opened the door to investigations, and controversies, into PC1's potential function as a novel G protein-coupled receptor (GPCR). Subsequent biochemical and cellular-based assays supported an ability of the PC1 C-tail to bind numerous members of the Gα protein family and to either inhibit or activate G protein-dependent pathways involved in the regulation of ion channel activity, transcription factor activation, and apoptosis. More recent work has demonstrated an essential role for PC1-mediated G protein regulation in preventing kidney cyst development; however, the mechanisms by which PC1 regulates G protein activity continue to be discovered. Similarities between PC1 and the adhesion class of 7-TM GPCRs, most notably a conserved GPCR proteolysis site (GPS) before the first TM domain, which undergoes autocatalyzed proteolytic cleavage, suggest potential mechanisms for PC1-mediated regulation of G protein signaling. This article reviews the evidence supporting GPCR-like functions of PC1 and their relevance to cystic disease, discusses the involvement of GPS cleavage and potential ligands in regulating PC1 GPCR function, and explores potential connections between PC1 GPCR-like activity and regulation of the channel properties of the polycystin receptor-channel complex.

3.
Proc Natl Acad Sci U S A ; 119(19): e2113786119, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35522707

ABSTRACT

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.


Subject(s)
Polycystic Kidney, Autosomal Dominant , TRPP Cation Channels , Female , Humans , Male , Mutation , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Signal Transduction , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism
4.
Front Cell Dev Biol ; 9: 661350, 2021.
Article in English | MEDLINE | ID: mdl-34095126

ABSTRACT

Primary cilia are small, antenna-like organelles that detect and transduce chemical and mechanical cues in the extracellular environment, regulating cell behavior and, in turn, tissue development and homeostasis. Primary cilia are assembled via intraflagellar transport (IFT), which traffics protein cargo bidirectionally along a microtubular axoneme. Ranging from 1 to 10 µm long, these organelles typically reach a characteristic length dependent on cell type, likely for optimum fulfillment of their specific roles. The importance of an optimal cilia length is underscored by the findings that perturbation of cilia length can be observed in a number of cilia-related diseases. Thus, elucidating mechanisms of cilia length regulation is important for understanding the pathobiology of ciliary diseases. Since cilia assembly/disassembly regulate cilia length, we review the roles of IFT in processes that affect cilia assembly/disassembly, including ciliary transport of structural and membrane proteins, ectocytosis, and tubulin posttranslational modification. Additionally, since the environment of a cell influences cilia length, we also review the various stimuli encountered by renal epithelia in healthy and diseased states that alter cilia length and IFT.

5.
Cell Signal ; 72: 109637, 2020 08.
Article in English | MEDLINE | ID: mdl-32305667

ABSTRACT

Polycystin-1, whose mutation is the most frequent cause of autosomal dominant polycystic kidney disease, is an extremely large and multi-faceted membrane protein whose primary or proximal cyst-preventing function remains undetermined. Accumulating evidence supports the idea that modulation of cellular signaling by heterotrimeric G proteins is a critical function of polycystin-1. The presence of a cis-autocatalyzed, G protein-coupled receptor (GPCR) proteolytic cleavage site, or GPS, in its extracellular N-terminal domain immediately preceding the first transmembrane domain is one of the notable conserved features of the polycystin-1-like protein family, and also of the family of cell adhesion GPCRs. Adhesion GPCRs are one of five families within the GPCR superfamily and are distinguished by a large N-terminal extracellular region consisting of multiple adhesion modules with a GPS-containing GAIN domain and bimodal functions in cell adhesion and signal transduction. Recent advances from studies of adhesion GPCRs provide a new paradigm for unraveling the mechanisms by which polycystin-1-associated G protein signaling contributes to the pathogenesis of polycystic kidney disease. This review highlights the structural and functional features shared by polycystin-1 and the adhesion GPCRs and discusses the implications of such similarities for our further understanding of the functions of this complicated protein.


Subject(s)
GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , TRPP Cation Channels/metabolism , Animals , Cell Adhesion , Humans , Models, Biological , Receptors, G-Protein-Coupled/chemistry , Signal Transduction , TRPP Cation Channels/chemistry
6.
Methods Cell Biol ; 153: 169-183, 2019.
Article in English | MEDLINE | ID: mdl-31395378

ABSTRACT

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.


Subject(s)
Embryo, Mammalian , Kidney , Organ Culture Techniques/methods , Polycystic Kidney Diseases/pathology , Animals , Culture Media/metabolism , Cyclic AMP/metabolism , Disease Models, Animal , Female , Humans , Intravital Microscopy/instrumentation , Intravital Microscopy/methods , Mice , Microdissection/instrumentation , Microdissection/methods , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods
7.
Hum Mol Genet ; 27(19): 3313-3324, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29931260

ABSTRACT

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.


Subject(s)
Heterotrimeric GTP-Binding Proteins/genetics , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics , Animals , CHO Cells , Calcium Channels/genetics , Cilia/genetics , Cilia/pathology , Cricetulus , Humans , Kidney/pathology , Mice , Mutation , Polycystic Kidney, Autosomal Dominant/pathology , Protein Domains/genetics , Signal Transduction
8.
Sci Rep ; 8(1): 3340, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29463793

ABSTRACT

Polycystic kidney disease (PKD) is a genetic disorder characterized by fluid-filled cysts in the kidney and liver that ultimately leads to end-stage renal disease. Currently there is no globally approved therapy for PKD. The Notch signaling pathway regulates cellular processes such as proliferation and de-differentiation, which are cellular hallmarks of PKD. Thus we hypothesized that the Notch pathway plays a critical role in PKD. Evaluation of protein expression of Notch signaling components in kidneys of Autosomal Recessive PKD (ARPKD) and Autosomal Dominant PKD (ADPKD) mouse models and of ADPKD patients revealed that Notch pathway members, particularly Notch3, were consistently upregulated or activated in cyst-lining epithelial cells. Notch3 expression correlated with rapidly growing cysts and co-localized with the proliferation marker, PCNA. Importantly, Notch inhibition significantly decreased forskolin-induced Notch3 activation and proliferation of primary human ADPKD cells, and significantly reduced cyst formation and growth of human ADPKD cells cultured in collagen gels. Thus our data indicate that Notch3 is aberrantly activated and facilitates epithelial cell proliferation in PKD, and that inhibition of Notch signaling may prevent cyst formation and growth.


Subject(s)
Gene Expression Regulation , Polycystic Kidney Diseases/pathology , Receptor, Notch3/analysis , Signal Transduction , Animals , Cell Proliferation , Disease Models, Animal , Epithelial Cells/pathology , Gene Expression Profiling , Humans , Mice , Middle Aged , Proliferating Cell Nuclear Antigen/analysis
9.
Results Probl Cell Differ ; 60: 281-321, 2017.
Article in English | MEDLINE | ID: mdl-28409350

ABSTRACT

Primary cilia are small, antenna-like structures that detect mechanical and chemical cues and transduce extracellular signals. While mammalian primary cilia were first reported in the late 1800s, scientific interest in these sensory organelles has burgeoned since the beginning of the twenty-first century with recognition that primary cilia are essential to human health. Among the most common clinical manifestations of ciliary dysfunction are renal cysts. The molecular mechanisms underlying renal cystogenesis are complex, involving multiple aberrant cellular processes and signaling pathways, while initiating molecular events remain undefined. Autosomal Dominant Polycystic Kidney Disease is the most common renal cystic disease, caused by disruption of polycystin-1 and polycystin-2 transmembrane proteins, which evidence suggests must localize to primary cilia for proper function. To understand how the absence of these proteins in primary cilia may be remediated, we review intracellular trafficking of polycystins to the primary cilium. We also examine the controversial mechanisms by which primary cilia transduce flow-mediated mechanical stress into intracellular calcium. Further, to better understand ciliary function in the kidney, we highlight the LKB1/AMPK, Wnt, and Hedgehog developmental signaling pathways mediated by primary cilia and misregulated in renal cystic disease.


Subject(s)
Cilia/pathology , Kidney Diseases, Cystic/physiopathology , Animals , Humans
10.
Mol Cell Biol ; 34(17): 3341-53, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24958103

ABSTRACT

Polycystin-1 (Pc1) cleavage at the G protein-coupled receptor (GPCR) proteolytic site (GPS) is required for normal kidney morphology in humans and mice. We found a complex pattern of endogenous Pc1 forms by GPS cleavage. GPS cleavage generates not only the heterodimeric cleaved full-length Pc1 (Pc1(cFL)) in which the N-terminal fragment (NTF) remains noncovalently associated with the C-terminal fragment (CTF) but also a novel (Pc1) form (Pc1(deN)) in which NTF becomes detached from CTF. Uncleaved Pc1 (Pc1(U)) resides primarily in the endoplasmic reticulum (ER), whereas both Pc1(cFL) and Pc1(deN) traffic through the secretory pathway in vivo. GPS cleavage is not a prerequisite, however, for Pc1 trafficking in vivo. Importantly, Pc1(deN) is predominantly found at the plasma membrane of renal epithelial cells. By functional genetic complementation with five Pkd1 mouse models, we discovered that CTF plays a crucial role in Pc1(deN) trafficking. Our studies support GPS cleavage as a critical regulatory mechanism of Pc1 biogenesis and trafficking for proper kidney development and homeostasis.


Subject(s)
Polycystic Kidney, Autosomal Dominant/metabolism , Receptors, G-Protein-Coupled/metabolism , TRPP Cation Channels/chemistry , TRPP Cation Channels/metabolism , Animals , Binding Sites , Biological Transport, Active , Cell Membrane/metabolism , Genetic Complementation Test , Golgi Apparatus/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Biological , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics
11.
Am J Pathol ; 184(7): 1957-66, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24815352

ABSTRACT

In polycystic kidney disease (PKD), renal parenchyma is destroyed by cysts, hypothesized to obstruct nephrons. A signature of unilateral ureteral obstruction, proximal tubular atrophy leads to formation of atubular glomeruli. To determine whether this process occurs in PKD, kidneys from pcy mice (moderately progressive PKD), kidneys from cpk mice (rapidly progressive PKD), and human autosomal dominant PKD were examined in early and late stages. Integrity of the glomerulotubular junction and proximal tubular mass were determined in sections stained with Lotus tetragonolobus lectin. Development of proximal tubular atrophy and atubular glomeruli was determined in serial sections of individual glomeruli. In pcy mice, most glomerulotubular junctions were normal at 20 weeks, but by 30 weeks, 56% were atrophic and 25% of glomeruli were atubular; glomerulotubular junction integrity decreased with increasing cyst area (r = 0.83, P < 0.05). In cpk mice, all glomerulotubular junctions were normal at 10 days, but by 19 days, 26% had become abnormal. In early-stage autosomal dominant PKD kidneys, 50% of glomeruli were atubular or attached to atrophic tubules; in advanced disease, 100% were abnormal. Thus, proximal tubular injury in cystic kidneys closely parallels that observed with ureteral obstruction. These findings support the hypothesis that, in renal cystic disorders, cyst-dependent obstruction of medullary and cortical tubules initiates a process culminating in widespread destruction of proximal convoluted tubules at the glomerulotubular junction.


Subject(s)
Kidney Glomerulus/pathology , Kidney Tubules, Proximal/pathology , Polycystic Kidney Diseases/pathology , Ureteral Obstruction/complications , Adult , Animals , Cysts , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL
12.
J Membr Biol ; 246(7): 581-90, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23784065

ABSTRACT

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.


Subject(s)
Drug Resistance/genetics , Gene Expression , Ouabain/pharmacology , Protein Interaction Domains and Motifs/genetics , TRPP Cation Channels/genetics , Animals , Anions/metabolism , Cell Line , Cell Proliferation/drug effects , Colforsin/pharmacology , ErbB Receptors/metabolism , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/metabolism , MAP Kinase Signaling System/drug effects , Mice , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , TRPP Cation Channels/chemistry
13.
Biochemistry ; 50(3): 349-55, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21142036

ABSTRACT

Polycystin-1 (PC1), the product of the polycystic kidney disease-1 (PKD1) gene, has a number of reported missense mutations whose pathogenicity is indeterminate. Previously, we utilized N-linked glycosylation reporter tags along with membrane insertion and topology assays to define the 11 membrane-spanning domains (I-XI) of PC1. In this report, we utilize glycosylation assays to determine whether two reported human polymorphisms/missense mutations within transmembrane (TM) domains VI and X affect the membrane topology of PC1. M3677T within TM VI had no effect on the topology of this TM domain as shown by the ability of two native N-linked glycosylation sites within the extracellular loop following TM VI to be glycosylated. In contrast, G4031D, within TM X, decreased the glycosylation of TM X reporter constructs, demonstrating that the substitution affected the C-terminal translocating activity of TM X. Furthermore, G4031D reduced the membrane association of TM X and XI together. These results suggest that G4031D affects the membrane insertion and topology of the C-terminal portion of polycystin-1 and represents a bona fide pathogenic mutation.


Subject(s)
Cell Membrane/metabolism , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism , Glycosylation , HEK293 Cells , Humans , Mutation, Missense , Polymorphism, Genetic , Protein Conformation , TRPP Cation Channels/chemistry
14.
J Biol Chem ; 285(50): 38818-31, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20923779

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Enzymologic , MAP Kinase Kinase Kinase 1/metabolism , Promoter Regions, Genetic , TRPP Cation Channels/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Animals , Base Sequence , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , Humans , Mice , Microscopy, Fluorescence/methods , Molecular Sequence Data , Mutagenesis , Oxidative Stress , Tumor Necrosis Factor-alpha/metabolism
15.
Am J Physiol Renal Physiol ; 295(6): F1845-54, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18922886

ABSTRACT

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.


Subject(s)
Promoter Regions, Genetic , TRPP Cation Channels/genetics , Transcriptional Activation/drug effects , Tretinoin/pharmacology , Tretinoin/physiology , Animals , Base Sequence , Cell Line , DNA Primers , Genes, Reporter , Humans , Kidney/embryology , Luciferases/genetics , Molecular Sequence Data , Plasmids , Receptors, Retinoic Acid/physiology , Retinoid X Receptors/physiology
16.
Am J Physiol Renal Physiol ; 295(6): F1725-34, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18829740

ABSTRACT

Polycystic kidney diseases (PKD) are inherited as autosomal dominant (ADPKD) or autosomal recessive (ARPKD) traits and are characterized by progressive enlargement of renal cysts. Aberrant cell proliferation is a key feature in the progression of PKD. Cux1 is a homeobox gene that is related to Drosophila cut and is the murine homolog of human CDP (CCAAT Displacement Protein). Cux1 represses the cyclin kinase inhibitors p21 and p27, and transgenic mice ectopically expressing Cux1 develop renal hyperplasia. However, Cux1 transgenic mice do not develop PKD. Here, we show that a 246 amino acid deletion in Cux1 accelerates PKD progression in cpk mice. Cystic kidneys isolated from 10-day-old cpk/Cux1 double mutant mice were significantly larger than kidneys from 10-day-old cpk mice. Moreover, renal function was significantly reduced in the Cux1 mutant cpk mice, compared with cpk mice. The mutant Cux1 protein was ectopically expressed in cyst-lining cells, where expression corresponded to increased cell proliferation and apoptosis, and a decrease in expression of the cyclin kinase inhibitors p27 and p21. While the mutant Cux1 protein altered PKD progression, kidneys from mice carrying the mutant Cux1 protein alone were phenotypically normal, suggesting the Cux1 mutation modifies PKD progression in cpk mice. During cell cycle progression, Cux1 is proteolytically processed by a nuclear isoform of the cysteine protease cathepsin-L. Analysis of the deleted sequences reveals that a cathepsin-L processing site in Cux1 is deleted. Moreover, nuclear cathepsin-L is significantly reduced in both human ADPKD cells and in Pkd1 null kidneys, corresponding to increased levels of Cux1 protein in the cystic cells and kidneys. These results suggest a mechanism in which reduced Cux1 processing by cathepsin-L results in the accumulation of Cux1, downregulation of p21/p27, and increased cell proliferation in PKD.


Subject(s)
Nuclear Proteins/deficiency , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/physiopathology , Animals , Cathepsin L , Cathepsins/genetics , Crosses, Genetic , Cysteine Endopeptidases/genetics , Disease Progression , Homeodomain Proteins/genetics , Humans , Mice , Mice, Knockout , Mutation , Nuclear Proteins/genetics , Organ Size , Polycystic Kidney Diseases/pathology , Polycystic Kidney, Autosomal Dominant/genetics , Repressor Proteins/genetics , Sequence Deletion
17.
J Am Soc Nephrol ; 17(12): 3424-37, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17108316

ABSTRACT

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.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Cyclic AMP/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Kidney Tubules/embryology , Polycystic Kidney Diseases/embryology , Sodium Potassium Chloride Symporter Inhibitors , Amides/pharmacology , Animals , Benzoates/pharmacology , Colforsin/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/physiopathology , Polycystic Kidney, Autosomal Dominant/etiology , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/physiopathology , Sodium-Potassium-Chloride Symporters/physiology , TRPP Cation Channels , Thiazolidines/pharmacology
18.
Biochem Biophys Res Commun ; 342(4): 1005-13, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16510125

ABSTRACT

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.


Subject(s)
Gelsolin/genetics , Promoter Regions, Genetic/genetics , Proteins/genetics , Proto-Oncogene Proteins c-ets/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Transcriptional Activation/genetics , Animals , Cells, Cultured , Drosophila , Gene Expression Regulation/genetics , Microfilament Proteins , TRPP Cation Channels , Trans-Activators
19.
Kidney Int ; 67(2): 432-42, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15673290

ABSTRACT

BACKGROUND: Cux-1 is a murine homeodomain protein that functions as a cell cycle-dependent transcriptional repressor in proliferating cells. Targets of Cux-1 repression include the cyclin kinase inhibitors p21 and p27. In the kidney, Cux-1 is spatially and temporally regulated, and ectopic expression of Cux-1 in transgenic mice results in renal hyperplasia. Previously, we observed that Cux-1 is deregulated in cystic kidneys from cpk mice. Recent studies have suggested a role for the cyclin kinase inhibitor p21 in the development of polycystic kidney disease (PKD) in mice lacking PKD1. METHODS: Since p21 is a target of transcriptional repression by Cux-1, we compared the expression of Cux-1 and p21 in kidneys from Pkd1 null and cpk mice by immunohistochemistry and Western blotting. We also evaluated apoptosis and the expression of the cyclin kinase inhibitor p27 in Pkd1 null and cpk mice by terminal deoxynucleotidal transferase (TdT)-mediated deoxyuridine triphosphate (dUTP) nick-end labeling (TUNEL) staining, immunohistochemistry, and Western blotting. RESULTS: In both early and late embryonic kidneys from Pkd1 null mice, Cux-1 was highly and ectopically expressed in normal-appearing tubule epithelium, interstitial cells, and in the epithelial cells lining the cysts, where it colocalized with proliferating cell nuclear antigen (PCNA). Increased Cux-1 expression in Pkd1 null kidneys was also associated with a decrease in p27 expression at late stages of cystogenesis. In cpk kidneys, Cux-1 was not up-regulated until late stages of cyst development. Moreover, in contrast to Pkd1 null kidneys, p21 and p27 were highly expressed in cpk kidneys. In late stages of cystogenesis, Cux-1 and p21 colocalized in cyst lining cells, which also showed a high incidence of apoptosis. CONCLUSION: These results suggest that cyst development in Pkd1 null mice and cpk mice proceeds through different mechanisms. In Pkd1 null mice, ectopic expression of Cux-1 is associated with increased cell proliferation. In contrast, in cpk mice, ectopic expression of Cux-1 is associated with apoptosis.


Subject(s)
Cell Cycle Proteins/analysis , Kidney/chemistry , Nuclear Proteins/analysis , Polycystic Kidney Diseases/metabolism , Proteins/physiology , Repressor Proteins/analysis , Animals , Apoptosis , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Homeodomain Proteins , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nuclear Proteins/physiology , Proliferating Cell Nuclear Antigen/analysis , Repressor Proteins/physiology , TRPP Cation Channels , Tumor Suppressor Proteins/analysis
20.
J Biol Chem ; 279(53): 55455-64, 2004 Dec 31.
Article in English | MEDLINE | ID: mdl-15466861

ABSTRACT

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.


Subject(s)
Calcineurin/metabolism , Proteins/physiology , Signal Transduction , Active Transport, Cell Nucleus , Animals , Blotting, Western , Boronic Acids/pharmacology , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels , Cell Line , Cell Nucleus/metabolism , Enzyme Activation , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Gadolinium/pharmacology , Genes, Reporter , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Inositol 1,4,5-Trisphosphate Receptors , Kidney/embryology , Kidney/metabolism , Lithium Chloride/pharmacology , Luciferases/metabolism , Macrocyclic Compounds , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Microscopy, Fluorescence , NFATC Transcription Factors , Oxazoles/pharmacology , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Pyrrolidinones/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ryanodine Receptor Calcium Release Channel/metabolism , TRPP Cation Channels , Time Factors , Tissue Distribution , Transfection , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...