Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 16(6): 1379-1403, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38684863

ABSTRACT

Polycystic kidney disease (PKD) is a genetic disorder characterized by bilateral cyst formation. We showed that PKD cells and kidneys display metabolic alterations, including the Warburg effect and glutaminolysis, sustained in vitro by the enzyme asparagine synthetase (ASNS). Here, we used antisense oligonucleotides (ASO) against Asns in orthologous and slowly progressive PKD murine models and show that treatment leads to a drastic reduction of total kidney volume (measured by MRI) and a prominent rescue of renal function in the mouse. Mechanistically, the upregulation of an ATF4-ASNS axis in PKD is driven by the amino acid response (AAR) branch of the integrated stress response (ISR). Metabolic profiling of PKD or control kidneys treated with Asns-ASO or Scr-ASO revealed major changes in the mutants, several of which are rescued by Asns silencing in vivo. Indeed, ASNS drives glutamine-dependent de novo pyrimidine synthesis and proliferation in cystic epithelia. Notably, while several metabolic pathways were completely corrected by Asns-ASO, glycolysis was only partially restored. Accordingly, combining the glycolytic inhibitor 2DG with Asns-ASO further improved efficacy. Our studies identify a new therapeutic target and novel metabolic vulnerabilities in PKD.


Subject(s)
Aspartate-Ammonia Ligase , Disease Models, Animal , Polycystic Kidney Diseases , Animals , Humans , Mice , Aspartate-Ammonia Ligase/metabolism , Aspartate-Ammonia Ligase/genetics , Aspartate-Ammonia Ligase/antagonists & inhibitors , Disease Progression , Kidney/pathology , Kidney/metabolism , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/drug therapy , Polycystic Kidney Diseases/pathology , Polycystic Kidney Diseases/genetics
2.
Nat Metab ; 5(3): 385-397, 2023 03.
Article in English | MEDLINE | ID: mdl-36879119

ABSTRACT

Depriving cells of nutrients triggers an energetic crisis, which is resolved by metabolic rewiring and organelle reorganization. Primary cilia are microtubule-based organelles at the cell surface, capable of integrating multiple metabolic and signalling cues, but their precise sensory function is not fully understood. Here we show that primary cilia respond to nutrient availability and adjust their length via glutamine-mediated anaplerosis facilitated by asparagine synthetase (ASNS). Nutrient deprivation causes cilia elongation, mediated by reduced mitochondrial function, ATP availability and AMPK activation independently of mTORC1. Of note, glutamine removal and replenishment is necessary and sufficient to induce ciliary elongation or retraction, respectively, under nutrient stress conditions both in vivo and in vitro by restoring mitochondrial anaplerosis via ASNS-dependent glutamate generation. Ift88-mutant cells lacking cilia show reduced glutamine-dependent mitochondrial anaplerosis during metabolic stress, due to reduced expression and activity of ASNS at the base of cilia. Our data indicate a role for cilia in responding to, and possibly sensing, cellular glutamine levels via ASNS during metabolic stress.


Subject(s)
Aspartate-Ammonia Ligase , Glutamine , Glutamine/metabolism , Aspartate-Ammonia Ligase/metabolism , Cilia/metabolism , Signal Transduction
3.
J Am Soc Nephrol ; 32(8): 1913-1932, 2021 08.
Article in English | MEDLINE | ID: mdl-34155062

ABSTRACT

BACKGROUND: In autosomal dominant polycystic kidney disease (ADPKD), cyst development and enlargement lead to ESKD. Macrophage recruitment and interstitial inflammation promote cyst growth. TWEAK is a TNF superfamily (TNFSF) cytokine that regulates inflammatory responses, cell proliferation, and cell death, and its receptor Fn14 (TNFRSF12a) is expressed in macrophage and nephron epithelia. METHODS: To evaluate the role of the TWEAK signaling pathway in cystic disease, we evaluated Fn14 expression in human and in an orthologous murine model of ADPKD. We also explored the cystic response to TWEAK signaling pathway activation and inhibition by peritoneal injection. RESULTS: Meta-analysis of published animal-model data of cystic disease reveals mRNA upregulation of several components of the TWEAK signaling pathway. We also observed that TWEAK and Fn14 were overexpressed in mouse ADPKD kidney cysts, and TWEAK was significantly high in urine and cystic fluid from patients with ADPKD. TWEAK administration induced cystogenesis and increased cystic growth, worsening the phenotype in a murine ADPKD model. Anti-TWEAK antibodies significantly slowed the progression of ADPKD, preserved renal function, and improved survival. Furthermore, the anti-TWEAK cystogenesis reduction is related to decreased cell proliferation-related MAPK signaling, decreased NF-κB pathway activation, a slight reduction of fibrosis and apoptosis, and an indirect decrease in macrophage recruitment. CONCLUSIONS: This study identifies the TWEAK signaling pathway as a new disease mechanism involved in cystogenesis and cystic growth and may lead to a new therapeutic approach in ADPKD.


Subject(s)
Cytokine TWEAK/metabolism , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , TWEAK Receptor/metabolism , Adult , Animals , Antibodies, Neutralizing/pharmacology , Apoptosis , Cell Proliferation/drug effects , Cysts/metabolism , Cysts/pathology , Cytokine TWEAK/antagonists & inhibitors , Cytokine TWEAK/genetics , Cytokine TWEAK/pharmacology , Disease Models, Animal , Disease Progression , Female , Fibrosis , Gene Expression , Humans , Macrophage Activation/drug effects , Macrophages , Male , Mice , Middle Aged , NF-kappa B/metabolism , Polycystic Kidney, Autosomal Dominant/physiopathology , Signal Transduction , TWEAK Receptor/genetics
4.
FASEB J ; 34(5): 6493-6507, 2020 05.
Article in English | MEDLINE | ID: mdl-32239723

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is a common monogenic disorder, characterized by bilateral renal cyst formation. Multiple pathways are de-regulated in cystic epithelia offering good opportunities for therapy. Others and we have previously reported that metabolic reprogramming, including alterations of the TCA cycle, are prominent features of ADPKD. Several lines of evidence suggest that mitochondrial impairment might be responsible for the metabolic alterations. Here, we performed morphologic and morphometric evaluation of mitochondria by TEM in an orthologous mouse model of PKD caused by mutations in the Pkd1 gene (Ksp-Cre;Pkd1flox/- ). Furthermore, we measured mitochondrial respiration by COX and SDH enzymatic activity in situ. We found several alterations including reduced mitochondrial mass, altered structure and fragmentation of the mitochondrial network in cystic epithelia of Ksp-Cre;Pkd1flox/- mice. At the molecular level, we found reduced expression of the pro-fusion proteins OPA1 and MFN1 and up-regulation of the pro-fission protein DRP1. Importantly, administration of Mdivi-1, which interferes with DRP1 rescuing mitochondrial fragmentation, significantly reduced kidney/body weight, cyst formation, and improved renal function in Ksp-Cre;Pkd1flox/- mice. Our data indicate that impaired mitochondrial structure and function play a role in disease progression, and that their improvement can significantly modify the course of the disease.


Subject(s)
Cysts/pathology , Disease Models, Animal , Mitochondria/pathology , Polycystic Kidney Diseases/pathology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/physiology , Animals , Cell Proliferation , Cysts/genetics , Cysts/metabolism , Disease Progression , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism
5.
Sci Rep ; 9(1): 16640, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31719603

ABSTRACT

Polycystin-1 (PC-1) and 2 (PC-2) are the products of the PKD1 and PKD2 genes, which are mutated in Autosomal Dominant Polycystic Kidney Disease (ADPKD). They form a receptor/channel complex that has been suggested to function as a mechanosensor, possibly activated by ciliary bending in the renal tubule, and resulting in calcium influx. This model has recently been challenged, leaving the question as to which mechanical stimuli activate the polycystins still open. Here, we used a SILAC/Mass-Spec approach to identify intracellular binding partners of tagged-endogenous PC-1 whereby we detected a class of interactors mediating regulation of cellular actomyosin contraction. Accordingly, using gain and loss-of-function cellular systems we found that PC-1 negatively regulates cellular contraction and YAP activation in response to extracellular stiffness. Thus, PC-1 enables cells to sense the rigidity of the extracellular milieu and to respond appropriately. Of note, in an orthologous murine model of PKD we found evidence of increased actomyosin contraction, leading to enhanced YAP nuclear translocation and transcriptional activity. Finally, we show that inhibition of ROCK-dependent actomyosin contraction by Fasudil reversed YAP activation and significantly improved disease progression, in line with recent studies. Our data suggest a possible direct role of PC-1 as a mechanosensor of extracellular stiffness.


Subject(s)
Actomyosin/physiology , TRPP Cation Channels/physiology , Animals , Disease Models, Animal , Dogs , Extracellular Space/metabolism , Fluorescent Antibody Technique , Gas Chromatography-Mass Spectrometry , Humans , Immunoprecipitation , Madin Darby Canine Kidney Cells , Mass Spectrometry , Mice , Mice, Inbred C57BL , Polycystic Kidney Diseases/metabolism , Real-Time Polymerase Chain Reaction
6.
Commun Biol ; 1: 194, 2018.
Article in English | MEDLINE | ID: mdl-30480096

ABSTRACT

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a genetic disorder caused by loss-of-function mutations in PKD1 or PKD2. Increased glycolysis is a prominent feature of the disease, but how it impacts on other metabolic pathways is unknown. Here, we present an analysis of mouse Pkd1 mutant cells and kidneys to investigate the metabolic reprogramming of this pathology. We show that loss of Pkd1 leads to profound metabolic changes that affect glycolysis, mitochondrial metabolism, and fatty acid synthesis (FAS). We find that Pkd1-mutant cells preferentially use glutamine to fuel the TCA cycle and to sustain FAS. Interfering with either glutamine uptake or FAS retards cell growth and survival. We also find that glutamine is diverted to asparagine via asparagine synthetase (ASNS). Transcriptional profiling of PKD1-mutant human kidneys confirmed these alterations. We find that silencing of Asns is lethal in Pkd1-mutant cells when combined with glucose deprivation, suggesting therapeutic approaches for ADPKD.

7.
Cell Rep ; 24(5): 1093-1104.e6, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30067967

ABSTRACT

Renal cell carcinomas (RCCs) are common cancers diagnosed in more than 350,000 people each year worldwide. Several pathways are de-regulated in RCCs, including mTORC1. However, how mTOR drives tumorigenesis in this context is unknown. The lack of faithful animal models has limited progress in understanding and targeting RCCs. Here, we generated a mouse model harboring the kidney-specific inactivation of Tsc1. These animals develop cysts that evolve into papillae, cystadenomas, and papillary carcinomas. Global profiling confirmed several metabolic derangements previously attributed to mTORC1. Notably, Tsc1 inactivation results in the accumulation of fumarate and in mTOR-dependent downregulation of the TCA cycle enzyme fumarate hydratase (FH). The re-expression of FH in cellular systems lacking Tsc1 partially rescued renal epithelial transformation. Importantly, the mTORC1-FH axis is likely conserved in human RCC specimens. We reveal a role of mTORC1 in renal tumorigenesis, which depends on the oncometabolite fumarate.


Subject(s)
Carcinoma, Renal Cell/metabolism , Fumarates/metabolism , Kidney Neoplasms/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Tuberous Sclerosis Complex 1 Protein/genetics , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cells, Cultured , Female , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Inbred C57BL , Up-Regulation
8.
Sci Rep ; 7(1): 1224, 2017 04 27.
Article in English | MEDLINE | ID: mdl-28450740

ABSTRACT

Protein synthesis is traditionally associated with specific cytoplasmic compartments. We now show that OFD1, a centrosomal/basal body protein, interacts with components of the Preinitiation complex of translation (PIC) and of the eukaryotic Initiation Factor (eIF)4F complex and modulates the translation of specific mRNA targets in the kidney. We demonstrate that OFD1 cooperates with the mRNA binding protein Bicc1 to functionally control the protein synthesis machinery at the centrosome where also the PIC and eIF4F components were shown to localize in mammalian cells. Interestingly, Ofd1 and Bicc1 are both involved in renal cystogenesis and selected targets were shown to accumulate in two models of inherited renal cystic disease. Our results suggest a possible role for the centrosome as a specialized station to modulate translation for specific functions of the nearby ciliary structures and may provide functional clues for the understanding of renal cystic disease.


Subject(s)
Centrosome/metabolism , Gene Expression Regulation , Protein Biosynthesis , Protein Interaction Mapping , Proteins/metabolism , RNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans
9.
Nat Commun ; 7: 10786, 2016 Mar 02.
Article in English | MEDLINE | ID: mdl-26931735

ABSTRACT

Previous studies report a cross-talk between the polycystic kidney disease (PKD) and tuberous sclerosis complex (TSC) genes. mTOR signalling is upregulated in PKD and rapamycin slows cyst expansion, whereas renal inactivation of the Tsc genes causes cysts. Here we identify a new interplay between the PKD and TSC genes, with important implications for the pathophysiology of both diseases. Kidney-specific inactivation of either Pkd1 or Tsc1 using an identical Cre (KspCre) results in aggressive or very mild PKD, respectively. Unexpectedly, we find that mTORC1 negatively regulates the biogenesis of polycystin-1 (PC-1) and trafficking of the PC-1/2 complex to cilia. Genetic interaction studies reveal an important role for PC-1 downregulation by mTORC1 in the cystogenesis of Tsc1 mutants. Our data potentially explain the severe renal manifestations of the TSC/PKD contiguous gene syndrome and open new perspectives for the use of mTOR inhibitors in autosomal dominant PKD caused by hypomorphic or missense PKD1 mutations.


Subject(s)
Cysts/pathology , Gene Expression Regulation/physiology , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , TRPP Cation Channels/metabolism , Tuberous Sclerosis/metabolism , Animals , Cilia , Down-Regulation , Gene Expression Regulation/drug effects , Gene Silencing , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/genetics , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , TRPP Cation Channels/genetics , Tuberous Sclerosis/genetics , Tuberous Sclerosis/pathology , Up-Regulation
10.
J Am Soc Nephrol ; 27(4): 1135-44, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26271513

ABSTRACT

Signaling from the primary cilium regulates kidney tubule development and cyst formation. However, the mechanism controlling targeting of ciliary components necessary for cilium morphogenesis and signaling is largely unknown. Here, we studied the function of class II phosphoinositide 3-kinase-C2α (PI3K-C2α) in renal tubule-derived inner medullary collecting duct 3 cells and show that PI3K-C2α resides at the recycling endosome compartment in proximity to the primary cilium base. In this subcellular location, PI3K-C2α controlled the activation of Rab8, a key mediator of cargo protein targeting to the primary cilium. Consistently, partial reduction of PI3K-C2α was sufficient to impair elongation of the cilium and the ciliary transport of polycystin-2, as well as to alter proliferation signals linked to polycystin activity. In agreement, heterozygous deletion of PI3K-C2α in mice induced cilium elongation defects in kidney tubules and predisposed animals to cyst development, either in genetic models of polycystin-1/2 reduction or in response to ischemia/reperfusion-induced renal damage. These results indicate that PI3K-C2α is required for the transport of ciliary components such as polycystin-2, and partial loss of this enzyme is sufficient to exacerbate the pathogenesis of cystic kidney disease.


Subject(s)
Cilia/physiology , Class II Phosphatidylinositol 3-Kinases/physiology , Kidney Diseases, Cystic , TRPP Cation Channels/physiology , Animals , Kidney Diseases, Cystic/etiology , Male , Mice , Signal Transduction
11.
J Am Soc Nephrol ; 27(7): 1958-69, 2016 07.
Article in English | MEDLINE | ID: mdl-26534924

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is an important cause of ESRD for which there exists no approved therapy in the United States. Defective glucose metabolism has been identified as a feature of ADPKD, and inhibition of glycolysis using glucose analogs ameliorates aggressive PKD in preclinical models. Here, we investigated the effects of chronic treatment with low doses of the glucose analog 2-deoxy-d-glucose (2DG) on ADPKD progression in orthologous and slowly progressive murine models created by inducible inactivation of the Pkd1 gene postnatally. As previously reported, early inactivation (postnatal days 11 and 12) of Pkd1 resulted in PKD developing within weeks, whereas late inactivation (postnatal days 25-28) resulted in PKD developing in months. Irrespective of the timing of Pkd1 gene inactivation, cystic kidneys showed enhanced uptake of (13)C-glucose and conversion to (13)C-lactate. Administration of 2DG restored normal renal levels of the phosphorylated forms of AMP-activated protein kinase and its target acetyl-CoA carboxylase. Furthermore, 2DG greatly retarded disease progression in both model systems, reducing the increase in total kidney volume and cystic index and markedly reducing CD45-positive cell infiltration. Notably, chronic administration of low doses (100 mg/kg 5 days per week) of 2DG did not result in any obvious sign of toxicity as assessed by analysis of brain and heart histology as well as behavioral tests. Our data provide proof of principle support for the use of 2DG as a therapeutic strategy in ADPKD.


Subject(s)
Deoxyglucose/therapeutic use , Polycystic Kidney, Autosomal Dominant/drug therapy , Animals , Disease Models, Animal , Disease Progression , Female , Male , Mice
12.
Nat Commun ; 5: 5482, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25405894

ABSTRACT

Primary cilia contain specific receptors and channel proteins that sense the extracellular milieu. Defective ciliary function causes ciliopathies such as autosomal dominant polycystic kidney disease (ADPKD). However, little is known about how large ciliary transmembrane proteins traffic to the cilia. Polycystin-1 (PC1) and -2 (PC2), the two ADPKD gene products, are large transmembrane proteins that co-localize to cilia where they act to control proper tubular diameter. Here we describe that PC1 and PC2 must interact and form a complex to reach the trans-Golgi network (TGN) for subsequent ciliary targeting. PC1 must also be proteolytically cleaved at a GPS site for this to occur. Using yeast two-hybrid screening coupled with a candidate approach, we identify a Rabep1/GGA1/Arl3-dependent ciliary targeting mechanism, whereby Rabep1 couples the polycystin complex to a GGA1/Arl3-based ciliary trafficking module at the TGN. This study provides novel insights into the ciliary trafficking mechanism of membrane proteins.


Subject(s)
ADP-Ribosylation Factors/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Cilia/metabolism , TRPP Cation Channels/metabolism , Vesicular Transport Proteins/metabolism , trans-Golgi Network/metabolism , ADP-Ribosylation Factors/genetics , Adaptor Proteins, Vesicular Transport/genetics , Animals , Cilia/genetics , Kidney/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Protein Binding , Protein Transport , TRPP Cation Channels/genetics , Vesicular Transport Proteins/genetics , trans-Golgi Network/genetics
13.
J Biol Chem ; 289(10): 6404-6414, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24459142

ABSTRACT

Mutations of the PKD1 and PKD2 genes, encoding polycystin-1 (PC1) and polycystin-2 (PC2), respectively, lead to autosomal dominant polycystic kidney disease. Interestingly, up-regulation or down-regulation of PKD1 or PKD2 leads to polycystic kidney disease in animal models, but their interrelations are not completely understood. We show here that full-length PC1 that interacts with PC2 via a C-terminal coiled-coil domain regulates PC2 expression in vivo and in vitro by down-regulating PC2 expression in a dose-dependent manner. Expression of the pathogenic mutant R4227X, which lacks the C-terminal coiled-coil domain, failed to down-regulate PC2 expression, suggesting that PC1-PC2 interaction is necessary for PC2 regulation. The proteasome and autophagy are two pathways that control protein degradation. Proteins that are not degraded by proteasomes precipitate in the cytoplasm and are transported via histone deacetylase 6 (HDAC6) toward the aggresomes. We found that HDAC6 binds to PC2 and that expression of full-length PC1 accelerates the transport of the HDAC6-PC2 complex toward aggresomes, whereas expression of the R4227X mutant fails to do so. Aggresomes are engulfed by autophagosomes, which then fuse with the lysosome for degradation; this process is also known as autophagy. We have now shown that PC1 overexpression leads to increased degradation of PC2 via autophagy. Interestingly, PC1 does not activate autophagy generally. Thus, we have now uncovered a new pathway suggesting that when PC1 is expressed, PC2 that is not bound to PC1 is directed to aggresomes and subsequently degraded via autophagy, a control mechanism that may play a role in autosomal dominant polycystic kidney disease pathogenesis.


Subject(s)
Autophagy , Phagosomes , TRPP Cation Channels/metabolism , Animals , Dogs , Down-Regulation , Histone Deacetylase 6 , Histone Deacetylases/metabolism , Kidney/metabolism , Madin Darby Canine Kidney Cells , Metabolic Networks and Pathways , Mice , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , TRPP Cation Channels/antagonists & inhibitors , TRPP Cation Channels/genetics
14.
Biochem Biophys Res Commun ; 444(4): 473-9, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24472557

ABSTRACT

The PKD1 gene is essential for a number of biological functions, and its loss-of-function causes autosomal dominant polycystic kidney disease (ADPKD). The gene is developmentally regulated and believed to play an essential role in renal development. Previous studies have shown that manipulating murine renal organ cultures with dominant-negative forms of the Pkd1 gene impaired ureteric bud (UB) branching. In the current study, we analyzed different stages of renal development in two distinct mouse models carrying either a null mutation or inactivation of the last two exons of Pkd1. Surprisingly, metanephric explants from Pkd1-deleted kidneys harvested at day E11.5 did not show defects of UB branching and elongation, estimated by cytokeratin staining on fixed tissues or by Hoxb7-GFP time-lapse imaging. However, renal explants from Pkd1-mutants isolated at day E14.5 showed impaired nephrogenesis. Notably, we observed cell migratory defects in the developing endothelial compartment. Previous studies had implicated the Pkd1 gene in controlling cell migration and collagen deposition through PI3 kinases. In line with these studies, our results show that wild-type explants treated with PI3-kinase inhibitors recapitulate the endothelial defects observed in Pkd1 mutants, whereas treatment with VEGF only partially rescued the defects. Our data are consistent with a role for the Pkd1 gene in the endothelium that may be required for proper nephrogenesis.


Subject(s)
Kidney Glomerulus/embryology , Kidney Glomerulus/physiopathology , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/physiopathology , TRPP Cation Channels/genetics , Animals , Cell Movement , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Deletion , Kidney Glomerulus/metabolism , Mice , Mutation , Organ Culture Techniques , Phosphoinositide-3 Kinase Inhibitors , TRPP Cation Channels/metabolism
15.
Nat Commun ; 4: 2658, 2013.
Article in English | MEDLINE | ID: mdl-24153433

ABSTRACT

Several organs, including the lungs and kidneys, are formed by epithelial tubes whose proper morphogenesis ensures correct function. This is best exemplified by the kidney, where defective establishment or maintenance of tubular diameter results in polycystic kidney disease, a common genetic disorder. Most polycystic kidney disease cases result from loss-of-function mutations in the PKD1 gene, encoding Polycystin-1, a large receptor of unknown function. Here we demonstrate that PC-1 has an essential role in the establishment of correct tubular diameter during nephron development. Polycystin-1 associates with Par3 favouring the assembly of a pro-polarizing Par3/aPKC complex and it regulates a programme of cell polarity important for oriented cell migration and for a convergent extension-like process during tubular morphogenesis. Par3 inactivation in the developing kidney results in defective convergent extension and tubular morphogenesis, and in renal cyst formation. Our data define Polycystin-1 as central to cell polarization and to epithelial tube morphogenesis and homeostasis.


Subject(s)
Cell Adhesion Molecules/genetics , Epithelial Cells/metabolism , Kidney Tubules/metabolism , Polycystic Kidney Diseases/genetics , Protein Kinase C-alpha/genetics , TRPP Cation Channels/genetics , Adaptor Proteins, Signal Transducing , Animals , Binding Sites , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins , Cell Movement , Cell Polarity , Embryo, Mammalian , Epithelial Cells/pathology , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Kidney Tubules/pathology , Male , Mice , Mice, Knockout , Morphogenesis/genetics , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology , Protein Binding , Protein Kinase C-alpha/metabolism , Protein Structure, Tertiary , Signal Transduction , TRPP Cation Channels/deficiency
16.
Nat Med ; 19(4): 488-93, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23524344

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is a common genetic disorder characterized by bilateral renal cyst formation. Recent identification of signaling cascades deregulated in ADPKD has led to the initiation of several clinical trials, but an approved therapy is still lacking. Using a metabolomic approach, we identify a pathogenic pathway in this disease that can be safely targeted for therapy. We show that mutation of PKD1 results in enhanced glycolysis in cells in a mouse model of PKD and in kidneys from humans with ADPKD. Glucose deprivation resulted in lower proliferation and higher apoptotic rates in PKD1-mutant cells than in nondeprived cells. Notably, two distinct PKD mouse models treated with 2-deoxyglucose (2DG), to inhibit glycolysis, had lower kidney weight, volume, cystic index and proliferation rates as compared to nontreated mice. These metabolic alterations depend on the extracellular signal-related kinase (ERK) pathway acting in a dual manner by inhibiting the liver kinase B1 (LKB1)-AMP-activated protein kinase (AMPK) axis on the one hand while activating the mTOR complex 1 (mTORC1)-glycolytic cascade on the other. Enhanced metabolic rates further inhibit AMPK. Forced activation of AMPK acts in a negative feedback loop, restoring normal ERK activity. Taken together, these data indicate that defective glucose metabolism is intimately involved in the pathobiology of ADPKD. Our findings provide a strong rationale for a new therapeutic strategy using existing drugs, either individually or in combination.


Subject(s)
Glucose/metabolism , Polycystic Kidney Diseases/metabolism , AMP-Activated Protein Kinase Kinases , Animals , Deoxyglucose/pharmacology , Disease Models, Animal , Glycolysis/drug effects , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/pathology , Protein Kinases/physiology , TRPP Cation Channels/genetics , TRPP Cation Channels/physiology
17.
PLoS One ; 4(9): e7137, 2009 Sep 23.
Article in English | MEDLINE | ID: mdl-19774080

ABSTRACT

Polycystin-1 (PC-1), the product of the PKD1 gene, mutated in the majority of cases of Autosomal Dominant Polycystic Kidney Disease (ADPKD), is a very large (approximately 520 kDa) plasma membrane receptor localized in several subcellular compartments including cell-cell/matrix junctions as well as cilia. While heterologous over-expression systems have allowed identification of several of the potential biological roles of this receptor, its precise function remains largely elusive. Studying PC-1 in vivo has been a challenging task due to its complexity and low expression levels. To overcome these limitations and facilitate the study of endogenous PC-1, we have inserted HA- or Myc-tag sequences into the Pkd1 locus by homologous recombination. Here, we show that our approach was successful in generating a fully functional and easily detectable endogenous PC-1. Characterization of PC-1 distribution in vivo showed that it is expressed ubiquitously and is developmentally-regulated in most tissues. Furthermore, our novel tool allowed us to investigate the role of PC-1 in brain, where the protein is abundantly expressed. Subcellular localization of PC-1 revealed strong and specific staining in ciliated ependymal and choroid plexus cells. Consistent with this distribution, we observed hydrocephalus formation both in the ubiquitous knock-out embryos and in newborn mice with conditional inactivation of the Pkd1 gene in the brain. Both choroid plexus and ependymal cilia were morphologically normal in these mice, suggesting a role for PC-1 in ciliary function or signalling in this compartment, rather than in ciliogenesis. We propose that the role of PC-1 in the brain cilia might be to prevent hydrocephalus, a previously unrecognized role for this receptor and one that might have important implications for other genetic or sporadic diseases.


Subject(s)
Brain/metabolism , Choroid Plexus/metabolism , Cilia/metabolism , Ependyma/metabolism , Hydrocephalus/metabolism , TRPP Cation Channels/deficiency , TRPP Cation Channels/genetics , Animals , Animals, Newborn , Epitopes/chemistry , Heterozygote , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL