Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nephrol Dial Transplant ; 38(10): 2266-2275, 2023 09 29.
Article in English | MEDLINE | ID: mdl-36914219

ABSTRACT

BACKGROUND: The epidermal growth factor receptor (EGFR) pathway is involved in kidney tissue repair and growth. Preclinical interventional data and scarce human data have suggested a role for this pathway in the pathophysiology of autosomal dominant polycystic kidney disease (ADPKD), while other data have suggested that its activation is causally linked to repair of damaged kidney tissue. We hypothesize that urinary EGFR ligands, as a reflection of EGFR activity, are associated with kidney function decline in ADPKD in the context of tissue repair following injury, and as the disease progresses as a sign of insufficient repair. METHODS: In the present study, we measured the EGFR ligands, EGF and heparin binding-EGF (HB-EGF), in 24-h urine samples of 301 ADPKD patients and 72 age- and sex-matched living kidney donors to dissect the role of the EGFR pathway in ADPKD. During a median follow-up of 2.5 years, the association of urinary EGFR ligand excretion with annual change in estimated glomerular filtration rate (eGFR) and height-adjusted total kidney volume in ADPKD patients was analyzed using mixed-models methods, and the expression of three closely related EGFR family receptors in ADPKD kidney tissue was investigated by immunohistochemistry. Additionally, the effect of reducing renal mass (after kidney donation), was assessed to investigate whether urinary EGF matches this reduction and thus reflects the amount of remaining healthy kidney tissue. RESULTS: At baseline, urinary HB-EGF did not differ between ADPKD patients and healthy controls (P = .6), whereas a lower urinary EGF excretion was observed in ADPKD patients [18.6 (11.8-27.8)] compared with healthy controls [51.0 (34.9-65.4) µg/24 h, P < .001]. Urinary EGF was positively associated with baseline eGFR (R = 0.54, P < .001) and a lower EGF was strongly associated with a more rapid GFR decline, even when adjusted for ADPKD severity markers (ß = 1.96, P < .001), whereas HB-EGF was not. Expression of the EGFR, but not other EGFR-related receptors, was observed in renal cysts but was absent in non-ADPKD kidney tissue. Finally, unilateral nephrectomy resulted in a decrease of 46.4 (-63.3 to -17.6) % in urinary EGF excretion, alongside a decrease of 35.2 ± 7.2% in eGFR and 36.8 ± 6.9% in measured GFR (mGFR), whereas maximal mGFR (measured after dopamine induced hyperperfusion) decreased by 46.1 ± 7.8% (all P < .001). CONCLUSIONS: Our data suggest that lower urinary EGF excretion may be a valuable novel predictor for kidney function decline in patients with ADPKD.


Subject(s)
Polycystic Kidney, Autosomal Dominant , Humans , Polycystic Kidney, Autosomal Dominant/complications , Heparin-binding EGF-like Growth Factor , Epidermal Growth Factor , Disease Progression , Kidney , Glomerular Filtration Rate , Patient Acuity
2.
Kidney Int ; 98(2): 404-419, 2020 08.
Article in English | MEDLINE | ID: mdl-32622526

ABSTRACT

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a major cause of end-stage kidney disease in man. The central role of cyclic adenosine monophosphate (cAMP) in ADPKD pathogenesis has been confirmed by numerous studies including positive clinical trial data. Here, we investigated the potential role of another major regulator of renal cAMP, prostaglandin E2 (PGE2), in modifying disease progression in ADPKD models using selective receptor modulators to all four PGE2 receptor subtypes (EP1-4). In 3D-culture model systems utilizing dog (MDCK) and patient-derived (UCL93, OX161-C1) kidney cell lines, PGE2 strikingly promoted cystogenesis and inhibited tubulogenesis by stimulating proliferation while reducing apoptosis. The effect of PGE2 on tubulogenesis and cystogenesis in 3D-culture was mimicked or abolished by selective EP2 and EP4 agonists or antagonists but not those specific to EP1 or EP3. In a Pkd1 mouse model (Pkd1nl/nl), kidney PGE2 and COX-2 expression were increased by two-fold at the peak of disease (week four). However, Pkd1nl/nl mice treated with selective EP2 (PF-04418948) or EP4 (ONO-AE3-208) antagonists from birth for three weeks had more severe cystic disease and fibrosis associated with increased cell proliferation and macrophage infiltration. A similar effect was observed for the EP4 antagonist ONO-AE3-208 in a second Pkd1 model (Pax8rtTA-TetO-Cre-Pkd1f/f). Thus, despite the positive effects of slowing cyst growth in vitro, the more complex effects of inhibiting EP2 or EP4 in vivo resulted in a worse outcome, possibly related to unexpected pro-inflammatory effects.


Subject(s)
Dinoprostone , Receptors, Prostaglandin E, EP2 Subtype , Animals , Cyclic AMP , Dogs , Humans , Inflammation/drug therapy , Kidney , Mice
3.
Kidney Int ; 98(2): 420-435, 2020 08.
Article in English | MEDLINE | ID: mdl-32622528

ABSTRACT

MicroRNAs (miRNAs) play an important role in regulating gene expression in health and disease but their role in modifying disease expression in Autosomal Dominant Polycystic Kidney Disease (ADPKD) remains uncertain. Here, we profiled human urinary exosome miRNA by global small RNA-sequencing in an initial discovery cohort of seven patients with ADPKD with early disease (eGFR over 60ml/min/1.73m2), nine with late disease (eGFR under 60ml/min/1.73m2), and compared their differential expression with six age and sex matched healthy controls. Two kidney-enriched candidate miRNA families were identified (miR-192/miR-194-2 and miR-30) and selected for confirmatory testing in a 60 patient validation cohort by quantitative polymerase chain reaction. We confirmed that miR-192-5p, miR-194-5p, miR-30a-5p, miR-30d-5p and miR-30e-5p were significantly downregulated in patient urine exosomes, in murine Pkd1 cystic kidneys and in human PKD1 cystic kidney tissue. All five miRNAs showed significant correlations with baseline eGFR and ultrasound-determined mean kidney length and improved the diagnostic performance (area under the curve) of mean kidney length for the rate of disease progression. Finally, inverse correlations of these two miRNA families with increased expression in their predicted target genes in patient PKD1 cystic tissue identified dysregulated pathways and transcriptional networks including novel interactions between miR-194-5p and two potentially relevant candidate genes, PIK3R1 and ANO1. Thus, our results identify a subset of urinary exosomal miRNAs that could serve as novel biomarkers of disease progression and suggest new therapeutic targets in ADPKD.


Subject(s)
Exosomes , MicroRNAs , Polycystic Kidney, Autosomal Dominant , Animals , Biomarkers , Exosomes/genetics , Gene Expression Profiling , Humans , Kidney , Mice , MicroRNAs/genetics , Polycystic Kidney, Autosomal Dominant/diagnosis , Polycystic Kidney, Autosomal Dominant/genetics
4.
JCI Insight ; 5(16)2020 08 20.
Article in English | MEDLINE | ID: mdl-32663194

ABSTRACT

Mutations in PKD1 (encoding for polycystin-1 [PC1]) are found in 80%-85% of patients with autosomal dominant polycystic kidney disease (ADPKD). We tested the hypothesis that changes in actin dynamics result from PKD1 mutations through dysregulation of compartmentalized centrosomal RhoA signaling mediated by specific RhoGAP (ARHGAP) proteins resulting in the complex cellular cystic phenotype. Initial studies revealed that the actin cytoskeleton was highly disorganized in cystic cells derived from patients with PKD1 and was associated with an increase in total and centrosomal active RhoA and ROCK signaling. Using cilia length as a phenotypic readout for centrosomal RhoA activity, we identified ARHGAP5, -29, and -35 as essential regulators of ciliation in normal human renal tubular cells. Importantly, a specific decrease in centrosomal ARHGAP35 was observed in PKD1-null cells using a centrosome-targeted proximity ligation assay and by dual immunofluorescence labeling. Finally, the ROCK inhibitor hydroxyfasudil reduced cyst expansion in both human PKD1 3D cyst assays and an inducible Pkd1 mouse model. In summary, we report a potentially novel interaction between PC1 and ARHGAP35 in the regulation of centrosomal RhoA activation and ROCK signaling. Targeting the RhoA/ROCK pathway inhibited cyst formation in vitro and in vivo, indicating its relevance to ADPKD pathogenesis and for developing new therapies to inhibit cyst initiation.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Repressor Proteins/metabolism , TRPP Cation Channels/metabolism , rhoA GTP-Binding Protein/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Actins/metabolism , Animals , Cell Line , Centrosome/metabolism , Cilia/metabolism , Disease Models, Animal , Guanine Nucleotide Exchange Factors/genetics , Humans , Mice, Transgenic , Mutation , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/metabolism , Protein Kinase C/genetics , Repressor Proteins/genetics , Signal Transduction , TRPP Cation Channels/genetics , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
5.
Cell Signal ; 72: 109644, 2020 08.
Article in English | MEDLINE | ID: mdl-32320857

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited cause of kidney failure and affects up to 12 million people worldwide. Germline mutations in two genes, PKD1 or PKD2, account for almost all patients with ADPKD. The ADPKD proteins, polycystin-1 (PC1) and polycystin-2 (PC2), are regulated by post-translational modifications (PTM), with phosphorylation, glycosylation and proteolytic cleavage being the best described changes. A few PTMs have been shown to regulate polycystin trafficking, signalling, localisation or stability and thus their physiological function. A key challenge for the future will be to elucidate the functional significance of all the individual PTMs reported to date. Finally, it is possible that site-specific mutations that disrupt PTM could contribute to cystogenesis although in the majority of cases, confirmatory evidence is awaited.


Subject(s)
Protein Processing, Post-Translational , TRPP Cation Channels/metabolism , Animals , Humans , Models, Biological , Phosphorylation , Proteome/metabolism , TRPP Cation Channels/chemistry
6.
J Am Soc Nephrol ; 28(10): 2973-2984, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28620080

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is caused by inactivating mutations in PKD1 (85%) or PKD2 (15%). The ADPKD proteins encoded by these genes, polycystin-1 (PC1) and polycystin-2 (PC2), form a plasma membrane receptor-ion channel complex. However, the mechanisms controlling the subcellular localization of PC1 and PC2 are poorly understood. Here, we investigated the involvement of the retromer complex, an ancient protein module initially discovered in yeast that regulates the retrieval, sorting, and retrograde transport of membrane receptors. Using yeast two-hybrid, biochemical, and cellular assays, we determined that PC2 binds two isoforms of the retromer-associated protein sorting nexin 3 (SNX3), including a novel isoform that binds PC2 in a direct manner. Knockdown of SNX3 or the core retromer protein VPS35 increased the surface expression of endogenous PC1 and PC2 in vitro and in vivo and increased Wnt-activated PC2-dependent whole-cell currents. These findings indicate that an SNX3-retromer complex regulates the surface expression and function of PC1 and PC2. Molecular targeting of proteins involved in the endosomal sorting of PC1 and PC2 could lead to new therapeutic approaches in ADPKD.


Subject(s)
Endocytosis , Sorting Nexins/metabolism , TRPP Cation Channels/metabolism , Animals , HEK293 Cells , HeLa Cells , Humans , Kidney Tubules/metabolism , Vesicular Transport Proteins/metabolism , Xenopus
7.
Am J Physiol Renal Physiol ; 312(4): F577-F588, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28077374

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is the fourth most common cause of end-stage renal disease. The disease course can be highly variable and treatment options are limited. To identify new therapeutic targets and prognostic biomarkers of disease, we conducted parallel discovery microarray profiling in normal and diseased human PKD1 cystic kidney cells. A total of 1,515 genes and 5 miRNA were differentially expressed by more than twofold in PKD1 cells. Functional enrichment analysis identified 30 dysregulated signaling pathways including the epidermal growth factor (EGF) receptor pathway. In this paper, we report that the EGF/ErbB family receptor ErbB4 is a major factor driving cyst growth in ADPKD. Expression of ErbB4 in vivo was increased in human ADPKD and Pkd1 cystic kidneys, both transcriptionally and posttranscriptionally by mir-193b-3p. Ligand-induced activation of ErbB4 drives cystic proliferation and expansion suggesting a pathogenic role in cystogenesis. Our results implicate ErbB4 activation as functionally relevant in ADPKD, both as a marker of disease activity and as a new therapeutic target in this major kidney disease.


Subject(s)
Cell Proliferation , Gene Expression Profiling/methods , Kidney/metabolism , Oligonucleotide Array Sequence Analysis , Polycystic Kidney, Autosomal Dominant/genetics , Receptor, ErbB-4/genetics , Animals , Case-Control Studies , Cell Proliferation/drug effects , Disease Models, Animal , Disease Progression , Genetic Association Studies , Genetic Markers , Genetic Predisposition to Disease , HEK293 Cells , Humans , Kidney/drug effects , Kidney/pathology , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Mutation , Neuregulin-1/pharmacology , Phenotype , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , RNA Interference , Receptor, ErbB-4/agonists , Receptor, ErbB-4/metabolism , Signal Transduction , TRPP Cation Channels/deficiency , TRPP Cation Channels/genetics , Transcriptional Activation , Transfection , Up-Regulation
8.
J Am Soc Nephrol ; 27(4): 1159-73, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26311459

ABSTRACT

Mutations in polycystin-1 (PC1) give rise to autosomal dominant polycystic kidney disease, an important and common cause of kidney failure. Despite its medical importance, the function of PC1 remains poorly understood. Here, we investigated the role of the intracellular polycystin-1, lipoxygenase, and α-toxin (PLAT) signature domain of PC1 using nuclear magnetic resonance, biochemical, cellular, and in vivo functional approaches. We found that the PLAT domain targets PC1 to the plasma membrane in polarized epithelial cells by a mechanism involving the selective binding of the PLAT domain to phosphatidylserine and L-α-phosphatidylinositol-4-phosphate (PI4P) enriched in the plasma membrane. This process is regulated by protein kinase A phosphorylation of the PLAT domain, which reduces PI4P binding and recruits ß-arrestins and the clathrin adaptor AP2 to trigger PC1 internalization. Our results reveal a physiological role for the PC1-PLAT domain in renal epithelial cells and suggest that phosphorylation-dependent internalization of PC1 is closely linked to its function in renal development and homeostasis.


Subject(s)
Lipoxygenase/physiology , TRPP Cation Channels/physiology , Humans , Lipoxygenase/genetics , Mutation , Protein Structure, Tertiary , TRPP Cation Channels/genetics
9.
J Biol Chem ; 290(39): 23888-96, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26269590

ABSTRACT

Polycystin-2 (PC2) is a TRP-type, Ca(2+)-permeable non-selective cation channel that plays an important role in Ca(2+) signaling in renal and non-renal cells. The effect(s) of the cAMP pathway and kinase mediated phosphorylation of PC2 seem to be relevant to PC2 trafficking and its interaction with polycystin-1. However, the role of PC2 phosphorylation in channel function is still poorly defined. Here we reconstituted apical membranes of term human syncytiotrophoblast (hST), containing endogenous PC2 (PC2hst), and in vitro translated channel protein (PC2iv). Addition of the catalytic subunit of PKA increased by 566% the spontaneous PC2hst channel activity in the presence of ATP. Interestingly, 8-Br-cAMP also stimulated spontaneous PC2hst channel activity in the absence of the exogenous kinase. Either stimulation was inhibited by addition of alkaline phosphatase, which in turn, was reversed by the phosphatase inhibitor vanadate. Neither maneuver modified the single channel conductance but instead increased channel mean open time. PKA directly phosphorylated PC2, which increased the mean open time but not the single channel conductance of the channel. PKA phosphorylation did not modify either R742X truncated or S829A-mutant PC2iv channel function. The data indicate that the cAMP pathway regulates PC2-mediated cation transport in the hST. The relevant PKA site for PC2 channel regulation centers on a single residue serine 829, in the carboxyl terminus.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Phosphorylation/physiology , Second Messenger Systems/physiology , TRPP Cation Channels/metabolism , Trophoblasts/metabolism , Amino Acid Substitution , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Humans , Ion Transport/physiology , Mutation, Missense , TRPP Cation Channels/genetics , Trophoblasts/cytology
10.
Hum Mol Genet ; 22(10): 1924-39, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23390129

ABSTRACT

Mutations in PKD1 (85%) or PKD2 (15%) account for almost all cases of autosomal dominant polycystic kidney disease (ADPKD). The ADPKD proteins, termed as polycystin-1 (PC1) and polycystin-2 (PC2), interact via their C-termini to form a receptor-ion channel complex whose function and regulation are not fully understood. Here, we report the first phosphorylated residue (Ser(829)) in PC2, whose dephosphorylation is mediated by PC1 binding through the recruitment of protein phosphatase-1 alpha (PP1α). Using a new phosphospecific antibody (pPC2) to this site, we demonstrate that Ser(829) is phosphorylated by Protein kinase A (PKA) but remains constitutively phosphorylated in cells and tissues lacking PC1. cAMP increased pSer(829) basolateral localization in MDCK cells in a time dependent manner and was essential for pronephric development in Xenopus embryos. When constitutively expressed, a complex phenotype associated with enhanced ATP-dependent ER Ca(2+) release and loss of growth suppression was observed in cycling cells. These results reveal a reciprocal functional link between PC1 and PC2 which is critically dependent on their interaction. Unopposed cAMP stimulated hyperphosphorylation of PC2 in the absence of functional PC1 could contribute to cyst initiation in PKD1 patients and represents a new molecular paradigm in understanding ADPKD pathogenesis.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Protein Phosphatase 1/metabolism , TRPP Cation Channels/metabolism , Animals , Cyclic AMP/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Dogs , HEK293 Cells , HeLa Cells , Humans , Madin Darby Canine Kidney Cells , Mice , Mice, Knockout , Mutation , Phosphorylation/physiology , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Protein Phosphatase 1/genetics , Protein Structure, Tertiary , TRPP Cation Channels/genetics , Xenopus laevis
11.
J Biol Chem ; 286(31): 27825-35, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21652693

ABSTRACT

Transglutaminase type 2 (TG2) catalyzes the formation of an ε-(γ-glutamyl)-lysine isopeptide bond between adjacent peptides or proteins including those of the extracellular matrix (ECM). Elevated extracellular TG2 leads to accelerated ECM deposition and reduced clearance that underlie tissue scarring and fibrosis. The extracellular trafficking of TG2 is crucial to its role in ECM homeostasis; however, the mechanism by which TG2 escapes the cell is unknown as it has no signal leader peptide and therefore cannot be transported classically. Understanding TG2 transport may highlight novel mechanisms to interfere with the extracellular function of TG2 as isoform-specific TG2 inhibitors remain elusive. Mammalian expression vectors were constructed containing domain deletions of TG2. These were transfected into three kidney tubular epithelial cell lines, and TG2 export was assessed to identify critical domains. Point mutation was then used to highlight specific sequences within the domain required for TG2 export. The removal of ß-sandwich domain prevented all TG2 export. Mutations of Asp(94) and Asp(97) within the N-terminal ß-sandwich domain were identified as crucial for TG2 externalization. These form part of a previously identified fibronectin binding domain ((88)WTATVVDQQDCTLSLQLTT(106)). However, siRNA knockdown of fibronectin failed to affect TG2 export. The sequence (88)WTATVVDQQDCTLSLQLTT(106) within the ß-sandwich domain of TG2 is critical to its export in tubular epithelial cell lines. The extracellular trafficking of TG2 is independent of fibronectin.


Subject(s)
GTP-Binding Proteins/metabolism , Kidney Tubules/metabolism , Transglutaminases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Culture Media, Conditioned , DNA Primers , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Fibronectins/chemistry , Fibronectins/genetics , Fibronectins/metabolism , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , Gene Knockdown Techniques , Kidney Tubules/cytology , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Glutamine gamma Glutamyltransferase 2 , Protein Transport , RNA, Small Interfering , Transfection , Transglutaminases/chemistry , Transglutaminases/genetics
12.
Am J Physiol Renal Physiol ; 300(6): F1375-84, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21429973

ABSTRACT

Thiazolidinediones have been reported to retard cystic disease in rodent models by uncertain mechanisms. We hypothesized that their major effect in retarding cystogenesis was through inhibiting cell proliferation or stimulating apoptosis. In the Madin-Darby canine kidney cell (MDCK) model, rosiglitazone inhibited cyst growth in a time- and dose-dependent manner and this was accompanied by a reduction in basal proliferation and an increase in apoptosis. Unexpectedly, we also observed a striking abnormality in lumen formation resulting in a characteristic multiple lumen or loss of lumen phenotype in treated cells at doses which did not inhibit cell proliferation. These changes were preceded by mislocalization of gp135 and Cdc42, misorientation of the mitotic spindle, and retardation in centrosome reorientation with later changes in primary cilia length and mislocalization of E-cadherin. Cdc42 activation was unaffected by rosiglitazone in monolayer culture but was profoundly inhibited in three-dimensional culture. MDCK cells stably expressing mutant Cdc42 showed a similar mislocalization of gp135 expression and multilumen phenotype in the absence of rosiglitazone. We conclude that rosiglitazone influences MDCK cyst growth by multiple mechanisms involving dosage-dependent effects on proliferation, spindle orientation, centrosome migration, and lumen formation. Correct spatial Cdc42 activation is critical for lumen formation, but the effect of rosiglitazone is likely to involve both Cdc42 and non-Cdc42 pathways.


Subject(s)
Cell Division/drug effects , Cell Polarity/drug effects , Kidney Diseases, Cystic/metabolism , Kidney/drug effects , Thiazolidinediones/pharmacology , Animals , Apoptosis , Cadherins/metabolism , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Dogs , Dose-Response Relationship, Drug , Kidney/metabolism , Rosiglitazone , Signal Transduction/drug effects , Time Factors
13.
Mol Biol Cell ; 21(22): 3853-65, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20881056

ABSTRACT

PKD2 is mutated in 15% of patients with autosomal dominant polycystic kidney disease. The PKD2 protein, polycystin-2 or TRPP2, is a nonselective Ca2+-permeable cation channel that has been shown to function at several locations, including primary cilia, basolateral membrane, and at the endoplasmic reticulum (ER). Nevertheless, the factors that regulate the channel activity of polycystin-2 are not well understood. Polycystin-2 has been shown previously to be regulated by phosphorylation at two serine residues (Ser812 and Ser76) with distinct functional consequences. Here, we report the identification of a previously unrecognized phosphorylation site within the polycystin-2 C terminus (Ser801), and we demonstrate that it is phosphorylated by protein kinase D. Phosphorylation at this site was significantly increased in response to serum and epidermal growth factor stimulation. In nonciliated Madin-Darby canine kidney I cells, inducible expression of polycystin-2 inhibited cell proliferation compared with wild-type cells. Mutagenesis at Ser801 abolished these effects and reduced ATP-stimulated Ca2+ release from ER stores. Finally, we show that a pathogenic mutation (S804N) within the consensus kinase recognition sequence abolished Ser801 phosphorylation. These results suggest that growth factor-stimulated, protein kinase D-mediated phosphorylation of polycystin-2 is essential for its ER channel function and links extracellular stimuli to its effects on cell growth and intracellular calcium regulation.


Subject(s)
Calcium Channels/metabolism , Cell Proliferation , Protein Kinase C/metabolism , TRPP Cation Channels/metabolism , Adenosine Triphosphate/pharmacology , Animals , Binding Sites/genetics , Blotting, Western , Calcium/metabolism , Calcium Channels/genetics , Cell Line , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Epidermal Growth Factor/pharmacology , Fluorescent Antibody Technique , HEK293 Cells , Humans , Mutation , Phosphorylation/drug effects , RNA Interference , Serine/genetics , Serine/metabolism , TRPP Cation Channels/genetics
14.
J Cell Sci ; 122(Pt 9): 1410-7, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19351715

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited human renal disease and is caused by mutations in two genes, PKD1 (85%) and PKD2 (15%). Cyst epithelial cells are characterised by a complex cellular phenotype including changes in proliferation, apoptosis, basement membrane composition and apicobasal polarity. Since polycystin 1 (PC1), the PKD1 protein, has been located in the basolateral membrane of kidney epithelial cells, we hypothesised that it might have a key role in mediating or stabilising cell-cell interactions. In non-ciliated L929 cells, stable or transient surface expression of the PC1 extracellular domain was sufficient to confer an adhesive phenotype and stimulate junction formation. In MDCK cells, we found that PC1 was recruited to the lateral membranes coincident with E-cadherin within 30 minutes after a ;calcium switch'. Recruitment of both proteins was significantly delayed when cells were treated with a PC1 blocking antibody raised to the PKD domains. Finally, PC1 and E-cadherin could be coimmunoprecipitated together from MDCK cells. We conclude that PC1 has a key role in initiating junction formation via initial homophilic interactions and facilitates junction assembly and the establishment of apicobasal polarity by E-cadherin recruitment.


Subject(s)
Cadherins/metabolism , Intercellular Junctions/metabolism , TRPP Cation Channels/metabolism , Animals , Cadherins/genetics , Calcium/metabolism , Cell Line , Cell Membrane/metabolism , Cell Polarity , Dogs , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , TRPP Cation Channels/genetics
15.
J Biol Chem ; 283(42): 28471-9, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18701462

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD), the most common inherited cause of kidney failure, is caused by mutations in either PKD1 (85%) or PKD2 (15%). The PKD2 protein, polycystin-2 (PC2 or TRPP2), is a member of the transient receptor potential (TRP) superfamily and functions as a non-selective calcium channel. PC2 has been found to form oligomers in native tissues suggesting that it may form functional homo- or heterotetramers with other subunits, similar to other TRP channels. Our experiments unexpectedly revealed that PC2 mutant proteins lacking the known C-terminal dimerization domain were still able to form oligomers and co-immunoprecipitate full-length PC2, implying the possible existence of a proximal dimerization domain. Using yeast two-hybrid and biochemical assays, we have mapped an alternative dimerization domain to the N terminus of PC2 (NT2-1-223, L224X). Functional characterization of this domain demonstrated that it was sufficient to induce cyst formation in zebrafish embryos and inhibit PC2 surface currents in mIMCD3 cells probably by a dominant-negative mechanism. In summary, we propose a model for PC2 assembly as a functional tetramer which depends on both C- and N-terminal dimerization domains. These results have significant implications for our understanding of PC2 function and disease pathogenesis in ADPKD and provide a new strategy for studying PC2 function.


Subject(s)
TRPP Cation Channels/chemistry , Animals , Dimerization , Electrophysiology/methods , Humans , Immunohistochemistry/methods , Models, Biological , Mutation , Plasmids/metabolism , Polycystic Kidney Diseases/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , TRPP Cation Channels/metabolism , Two-Hybrid System Techniques , Zebrafish
16.
EMBO J ; 27(9): 1345-56, 2008 May 07.
Article in English | MEDLINE | ID: mdl-18388856

ABSTRACT

The TRPP2 cation channel is directly responsible for approximately 15% of all cases of autosomal dominant polycystic kidney disease. However, the mechanisms underlying fundamental properties of TRPP2 regulation, such as channel gating and activation, are unknown. We have shown that TRPP2 was activated by EGF and physically interacted with the mammalian diaphanous-related formin 1 (mDia1), a downstream effector of RhoA. Now, we show that mDia1 regulates TRPP2 by specifically blocking its activity at negative but not positive potentials. The voltage-dependent unblock of TRPP2 by mDia1 at positive potentials is mediated through RhoA-induced molecular switching of mDia1 from its autoinhibited state at negative potentials to its activated state at positive potentials. Under physiological resting potentials, EGF activates TRPP2 by releasing the mDia1-dependent block through the activation of RhoA. Our data reveal a new role of mDia1 in the regulation of ion channels and suggest a molecular basis for the voltage-dependent gating of TRP channels.


Subject(s)
Carrier Proteins/metabolism , Ion Channel Gating/physiology , TRPP Cation Channels/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Line , Cell Membrane/metabolism , Electrophysiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/physiology , Ion Channel Gating/genetics , Kidney/cytology , Membrane Potentials/physiology , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/physiopathology , Signal Transduction/physiology , Transfection , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/physiology , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
17.
Hum Mol Genet ; 15(9): 1465-73, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16551655

ABSTRACT

PKD2 is mutated in 15% of patients with autosomal dominant polycystic kidney disease. Polycystin-2 (PC2), the PKD2 protein, is a non-selective Ca(2+)-permeable cation channel which may function at the cell surface and ER. Nevertheless, the factors that regulate the dynamic translocation of PC2 between the ER and other compartments are not well understood. Constitutive phosphorylation of PC2 at a single C-terminal site (Ser(812)) has been previously reported. As we were unable to abolish phospholabelling of PC2 in HEK293 cells by site-directed mutagenesis of Ser(812) or all five predicted phosphorylation sites in the C-terminus, we hypothesized that PC2 could also be phosphorylated at the N-terminus. In this paper, we report the identification of a new phosphorylation site for PC2 within its N-terminal domain (Ser(76)) and demonstrate that this residue is phosphorylated by glycogen synthase kinase 3 (GSK3). The consensus recognition sequence for GSK3 (Ser(76)/Ser(80)) is evolutionarily conserved down to lower vertebrates. In the presence of specific GSK3 inhibitors, the lateral plasma membrane pool of endogenous PC2 redistributes into an intracellular compartment in MDCK cells without any change in primary cilia localization. Finally, co-injection of wild-type but not a S76A/S80A mutant PKD2 capped mRNA could rescue the cystic phenotype induced by an antisense morpholino oligonucleotide to pkd2 in zebrafish pronephric kidney. We conclude that surface localization of PC2 is regulated by phosphorylation at a unique GSK3 site in its N-terminal domain in vivo and in vitro. This site is functionally significant for the maintenance of normal glomerular and tubular morphology.


Subject(s)
Glycogen Synthase Kinase 3/physiology , Peptide Fragments/metabolism , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/chemistry , TRPP Cation Channels/metabolism , Animals , Cell Line , Dogs , Humans , Phosphorylation , Polycystic Kidney, Autosomal Dominant/enzymology , Protein Structure, Tertiary , Zebrafish
18.
J Am Soc Nephrol ; 14(7): 1804-15, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12819240

ABSTRACT

The PKD1 protein, polycystin-1, is a large transmembrane protein of uncertain function and topology. To study the putative functions of polycystin-1, conditionally immortalized kidney cells transgenic for PKD1 were generated and an interaction between transgenic polycystin-1 and endogenous polycystin-2 has been recently demonstrated in these cells. This study provides the first functional evidence that transgenic polycystin-1 directly mediates cell-cell adhesion. In non-permeabilized cells, polycystin-1 localized to the lateral cell borders with N-terminal antibodies but not with a C-terminal antibody; there was a clear difference in surface intensity between transgenic and non-transgenic cells. Compared with non-transgenic cells, transgenic cells showed a dramatic increase in resistance to the disruptive effect of a polycystin-1 antibody raised to the PKD domains of polycystin-1 (IgPKD) in both cell adhesion and cell aggregation assays. The differential effect on cell adhesion between transgenic and non-transgenic cells could be reproduced using recombinant fusion proteins encoding non-overlapping regions of the IgPKD domains. In contrast, antibodies raised to other extracellular domains of polycystin-1 had no effect on cell adhesion. Finally, the specificity of this finding was confirmed by the lack of effect of IgPKD antibody on cell adhesion in a PKD1 cystic cell line deficient in polycystin-1. These results demonstrate that one of the primary functions of polycystin-1 is to mediate cell-cell adhesion in renal epithelial cells, probably via homophilic or heterophilic interactions of the PKD domains. Disruption of cell-cell adhesion during tubular morphogenesis may be an early initiating event for cyst formation in ADPKD.


Subject(s)
Proteins/genetics , Proteins/physiology , Animals , Apoptosis , Cell Adhesion , Cell Aggregation , Cell Line , Cell Survival , Glutathione Transferase/metabolism , Humans , Immunoblotting , In Situ Nick-End Labeling , Kidney/cytology , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Fluorescence , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , TRPP Cation Channels
19.
J Biol Chem ; 277(23): 20763-73, 2002 Jun 07.
Article in English | MEDLINE | ID: mdl-11901144

ABSTRACT

The functions of the two proteins defective in autosomal dominant polycystic kidney disease, polycystin-1 and polycystin-2, have not been fully clarified, but it has been hypothesized that they may heterodimerize to form a "polycystin complex" involved in cell adhesion. In this paper, we demonstrate for the first time the existence of a native polycystin complex in mouse kidney tubular cells transgenic for PKD1, non-transgenic kidney cells, and normal adult human kidney. Polycystin-1 is heavily N-glycosylated, and several glycosylated forms of polycystin-1 differing in their sensitivity to endoglycosidase H (Endo H) were found; in contrast, native polycystin-2 was fully Endo H-sensitive. Using highly specific antibodies to both proteins, we show that polycystin-2 associates selectively with two species of full-length polycystin-1, one Endo H-sensitive and the other Endo H-resistant; importantly, the latter could be further enriched in plasma membrane fractions and co-immunoprecipitated with polycystin-2. Finally, a subpopulation of this complex co-localized to the lateral cell borders of PKD1 transgenic kidney cells. These results demonstrate that polycystin-1 and polycystin-2 interact in vivo to form a stable heterodimeric complex and suggest that disruption of this complex is likely to be of primary relevance to the pathogenesis of cyst formation in autosomal dominant polycystic kidney disease.


Subject(s)
Membrane Proteins/metabolism , Proteins/metabolism , Animals , Cell Membrane/metabolism , Humans , Kidney/metabolism , Membrane Proteins/genetics , Mice , Mice, Transgenic , Precipitin Tests , Protein Binding , Proteins/genetics , TRPP Cation Channels
SELECTION OF CITATIONS
SEARCH DETAIL