Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
PLoS One ; 18(8): e0289778, 2023.
Article in English | MEDLINE | ID: mdl-37540694

ABSTRACT

PKD1 is the most commonly mutated gene causing autosomal dominant polycystic kidney disease (ADPKD). It encodes Polycystin-1 (PC1), a putative membrane protein that undergoes a set of incompletely characterized post-transcriptional cleavage steps and has been reported to localize in multiple subcellular locations, including the primary cilium and mitochondria. However, direct visualization of PC1 and detailed characterization of its binding partners remain challenging. We now report a new mouse model with HA epitopes and eGFP knocked-in frame into the endogenous mouse Pkd1 gene by CRISPR/Cas9. Using this model, we sought to visualize endogenous PC1-eGFP and performed affinity-purification mass spectrometry (AP-MS) and network analyses. We show that the modified Pkd1 allele is fully functional but the eGFP-tagged protein cannot be detected without signal amplification by secondary antibodies. Using nanobody-coupled beads and large quantities of tissue, AP-MS identified an in vivo PC1 interactome, which is enriched for mitochondrial proteins and components of metabolic pathways. These studies suggest this mouse model and interactome data will be useful to understand PC1 function, but that new methods and brighter tags will be required to track endogenous PC1.


Subject(s)
Polycystic Kidney Diseases , Polycystic Kidney, Autosomal Dominant , Mice , Animals , TRPP Cation Channels/chemistry , Polycystic Kidney, Autosomal Dominant/genetics , Disease Models, Animal
3.
Adv Kidney Dis Health ; 30(3): 209-219, 2023 05.
Article in English | MEDLINE | ID: mdl-37088523

ABSTRACT

Autosomal dominant polycystic kidney disease is the most common inherited cause of end-stage kidney disease worldwide. Most cases result from mutation of either of 2 genes, PKD1 and PKD2, which encode proteins that form a probable receptor/channel complex. Studies suggest that a loss of function of the complex below an indeterminate threshold triggers cyst initiation, which ultimately results in dysregulation of multiple metabolic processes and downstream pathways and subsequent cyst growth. Noncell autonomous factors may also promote cyst growth. In this report, we focus primarily on the process of early cyst formation and factors that contribute to its variability with brief consideration of how new studies suggest this process may be reversible.


Subject(s)
Polycystic Kidney Diseases , Polycystic Kidney, Autosomal Dominant , Humans , TRPP Cation Channels/genetics , Polycystic Kidney Diseases/genetics , Mutation , Polycystic Kidney, Autosomal Dominant/genetics
5.
Kidney360 ; 2(10): 1576-1591, 2021 10 28.
Article in English | MEDLINE | ID: mdl-35372986

ABSTRACT

Background: Multiple studies of tissue and cell samples from patients and preclinical models of autosomal dominant polycystic kidney disease report abnormal mitochondrial function and morphology and suggest metabolic reprogramming is an intrinsic feature of this disease. Peroxisomes interact with mitochondria physically and functionally, and congenital peroxisome biogenesis disorders can cause various phenotypes, including mitochondrial defects, metabolic abnormalities, and renal cysts. We hypothesized that a peroxisomal defect might contribute to the metabolic and mitochondrial impairments observed in autosomal dominant polycystic kidney disease. Methods: Using control and Pkd1-/- kidney epithelial cells, we investigated peroxisome abundance, biogenesis, and morphology by immunoblotting, immunofluorescence, and live cell imaging of peroxisome-related proteins and assayed peroxisomal specific ß-oxidation. We further analyzed fatty acid composition by mass spectrometry in kidneys of Pkd1fl/fl;Ksp-Cre mice. We also evaluated peroxisome lipid metabolism in published metabolomics datasets of Pkd1 mutant cells and kidneys. Lastly, we investigated if the C terminus or full-length polycystin-1 colocalize with peroxisome markers by imaging studies. Results: Peroxisome abundance, morphology, and peroxisome-related protein expression in Pkd1-/- cells were normal, suggesting preserved peroxisome biogenesis. Peroxisomal ß-oxidation was not impaired in Pkd1-/- cells, and there was no obvious accumulation of very-long-chain fatty acids in kidneys of mutant mice. Reanalysis of published datasets provide little evidence of peroxisomal abnormalities in independent sets of Pkd1 mutant cells and cystic kidneys, and provide further evidence of mitochondrial fatty acid oxidation defects. Imaging studies with either full-length polycystin-1 or its C terminus, a fragment previously shown to go to the mitochondria, showed minimal colocalization with peroxisome markers restricted to putative mitochondrion-peroxisome contact sites. Conclusions: Our studies showed that loss of Pkd1 does not disrupt peroxisome biogenesis nor peroxisome-dependent fatty acid metabolism.


Subject(s)
Polycystic Kidney Diseases , Polycystic Kidney, Autosomal Dominant , Protein Kinase C/metabolism , Animals , Humans , Lipid Metabolism/genetics , Mice , Mutation , Peroxisomes/metabolism , Polycystic Kidney Diseases/genetics , Polycystic Kidney, Autosomal Dominant/genetics
6.
Cell Signal ; 74: 109701, 2020 10.
Article in English | MEDLINE | ID: mdl-32649993

ABSTRACT

Systems-based, agnostic approaches focusing on transcriptomics data have been employed to understand the pathogenesis of polycystic kidney diseases (PKD). While multiple signaling pathways, including Wnt, mTOR and G-protein-coupled receptors, have been implicated in late stages of disease, there were few insights into the transcriptional cascade immediately downstream of Pkd1 inactivation. One of the consistent findings has been transcriptional evidence of dysregulated metabolic and cytoskeleton remodeling pathways. Recent technical developments, including bulk and single-cell RNA sequencing technologies and spatial transcriptomics, offer new angles to investigate PKD. In this article, we review what has been learned based on transcriptional approaches and consider future opportunities.


Subject(s)
Polycystic Kidney Diseases/metabolism , Transcriptome , Animals , Gene Expression Profiling , Humans , TRPP Cation Channels/metabolism
7.
Nat Rev Nephrol ; 15(12): 735-749, 2019 12.
Article in English | MEDLINE | ID: mdl-31488901

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) affects an estimated 1 in 1,000 people and slowly progresses to end-stage renal disease (ESRD) in about half of these individuals. Tolvaptan, a vasopressin 2 receptor blocker, has been approved by regulatory authorities in many countries as a therapy to slow cyst growth, but additional treatments that target dysregulated signalling pathways in cystic kidney and liver are needed. Metabolic reprogramming is a prominent feature of cystic cells and a potentially important contributor to the pathophysiology of ADPKD. A number of pathways previously implicated in the pathogenesis of the disease, such as dysregulated mTOR and primary ciliary signalling, have roles in metabolic regulation and may exert their effects through this mechanism. Some of these pathways are amenable to manipulation through dietary modifications or drug therapies. Studies suggest that polycystin-1 and polycystin-2, which are encoded by PKD1 and PKD2, respectively (the genes that are mutated in >99% of patients with ADPKD), may in part affect cellular metabolism through direct effects on mitochondrial function. Mitochondrial dysfunction could alter the redox state and cellular levels of acetyl-CoA, resulting in altered histone acetylation, gene expression, cytoskeletal architecture and response to cellular stress, and in an immunological response that further promotes cyst growth and fibrosis.


Subject(s)
Polycystic Kidney Diseases/metabolism , Animals , Humans , Kidney/metabolism , Kidney/physiopathology , Metabolic Networks and Pathways , Polycystic Kidney Diseases/etiology , Polycystic Kidney Diseases/pathology
8.
Sci Rep ; 8(1): 2743, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29426897

ABSTRACT

Recent studies have reported intrinsic metabolic reprogramming in Pkd1 knock-out cells, implicating dysregulated cellular metabolism in the pathogenesis of polycystic kidney disease. However, the exact nature of the metabolic changes and their underlying cause remains controversial. We show herein that Pkd1 k o /ko renal epithelial cells have impaired fatty acid utilization, abnormal mitochondrial morphology and function, and that mitochondria in kidneys of ADPKD patients have morphological alterations. We further show that a C-terminal cleavage product of polycystin-1 (CTT) translocates to the mitochondria matrix and that expression of CTT in Pkd1 ko/ko cells rescues some of the mitochondrial phenotypes. Using Drosophila to model in vivo effects, we find that transgenic expression of mouse CTT results in decreased viability and exercise endurance but increased CO2 production, consistent with altered mitochondrial function. Our results suggest that PC1 may play a direct role in regulating mitochondrial function and cellular metabolism and provide a framework to understand how impaired mitochondrial function could be linked to the regulation of tubular diameter in both physiological and pathological conditions.


Subject(s)
Kidney , Mitochondria , Mitochondrial Proteins/metabolism , Polycystic Kidney, Autosomal Dominant/metabolism , Proteolysis , TRPP Cation Channels/metabolism , Aged , Animals , Animals, Genetically Modified , Dogs , Drosophila melanogaster , Embryo, Mammalian , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fatty Acids/metabolism , Gene Knockdown Techniques , Humans , Kidney/metabolism , Kidney/pathology , Madin Darby Canine Kidney Cells , Male , Mice , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/genetics , TRPP Cation Channels/genetics
9.
J Womens Health (Larchmt) ; 27(1): 115-120, 2018 01.
Article in English | MEDLINE | ID: mdl-29341869

ABSTRACT

Sex and gender are critical contributors to overall health and disease, and considering both in research informs the development of prevention strategies and treatment interventions for both men and women. The National Institutes of Health (NIH) Office of Research on Women's Health sponsored a preconference workshop on this topic at the 24th Annual Women's Health Congress, which was held in Crystal City, VA, in April 2016. The workshop featured presentations by NIH intramural and extramural scientists who presented data on a variety of topics including polycystic kidney disease, vaccine protection, depression, drug addiction, and cardiovascular disease. In this publication, we discuss the major points of each presentation and demonstrate the importance of considering sex and gender in biomedical research.


Subject(s)
Biomedical Research , Congresses as Topic , Health Status Disparities , Women's Health , Cardiovascular Diseases , Depressive Disorder, Major , Female , Humans , National Institutes of Health (U.S.) , Sex Factors , Substance-Related Disorders , United States
10.
Sci Rep ; 7(1): 7733, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28798345

ABSTRACT

Autosomal recessive polycystic kidney disease (ARPKD) is an important childhood nephropathy, occurring 1 in 20,000 live births. The major clinical phenotypes are expressed in the kidney with dilatation of the collecting ducts, systemic hypertension, and progressive renal insufficiency, and in the liver with biliary dysgenesis, portal tract fibrosis, and portal hypertension. The systemic hypertension has been attributed to enhanced distal sodium reabsorption in the kidney, the structural defects have been ascribed to altered cellular morphology, and fibrosis to increased TGF-ß signaling in the kidney and biliary tract, respectively. The pathogenic mechanisms underlying these abnormalities have not been determined. In the current report, we find that disrupting PKHD1 results in altered sub-cellular localization and function of the C2-WWW-HECT domain E3 family of ligases regulating these processes. We also demonstrate altered activity of RhoA and increased TGF-ß signaling and ENaC activity. Linking these phenomena, we found that vesicles containing the PKHD1/Pkhd1 gene product, FPC, also contain the NEDD4 ubiquitin ligase interacting protein, NDFIP2, which interacts with multiple members of the C2-WWW-HECT domain E3 family of ligases. Our results provide a mechanistic explanation for both the cellular effects and in vivo phenotypic abnormalities in mice and humans that result from Pkhd1/PKHD1 mutation.


Subject(s)
Nedd4 Ubiquitin Protein Ligases/metabolism , Polycystic Kidney, Autosomal Recessive/genetics , Polycystic Kidney, Autosomal Recessive/metabolism , Receptors, Cell Surface/deficiency , Animals , Biomarkers , Cell Line , Disease Models, Animal , Enzyme Activation , Gene Expression , Humans , Intracellular Space/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Mutation , Polycystic Kidney, Autosomal Recessive/pathology , Protein Transport , Rats , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction , rho GTP-Binding Proteins/metabolism
11.
Kidney Int ; 92(5): 1130-1144, 2017 11.
Article in English | MEDLINE | ID: mdl-28729032

ABSTRACT

Autosomal recessive polycystic kidney disease (OMIM 263200) is a serious condition of the kidney and liver caused by mutations in a single gene, PKHD1. This gene encodes fibrocystin/polyductin (FPC, PD1), a large protein shown by in vitro studies to undergo Notch-like processing. Its cytoplasmic tail, reported to include a ciliary targeting sequence, a nuclear localization signal, and a polycystin-2 binding domain, is thought to traffic to the nucleus after cleavage. We now report a novel mouse line with a triple HA-epitope "knocked-in" to the C-terminus along with lox P sites flanking exon 67, which encodes most of the C-terminus (Pkhd1Flox67HA). The triple HA-epitope has no functional effect as assayed by phenotype and allows in vivo tracking of Fibrocystin. We used the HA tag to identify previously predicted Fibrocystin cleavage products in tissue. In addition, we found that Polycystin-2 fails to co-precipitate with Fibrocystin in kidney samples. Immunofluorescence studies with anti-HA antibodies demonstrate that Fibrocystin is primarily present in a sub-apical location the in kidney, biliary duct, and pancreatic ducts, partially overlapping with the Golgi. In contrast to previous studies, the endogenous protein in the primary cilia was not detectable in mouse tissues. After Cre-mediated deletion, homozygous Pkhd1Δ67 mice are completely normal. Thus, Pkhd1Flox67HA is a valid model to track Pkhd1-derived products containing the C-terminus. Significantly, exon 67 containing the nuclear localization signal and the polycystin-2 binding domain is not essential for Fibrocystin function in our model.


Subject(s)
Kidney/metabolism , Polycystic Kidney, Autosomal Recessive/genetics , Protein Domains/genetics , Receptors, Cell Surface/genetics , TRPP Cation Channels/metabolism , Animals , Cilia/metabolism , Disease Models, Animal , Epitopes/genetics , Exons/genetics , Female , Fluorescent Antibody Technique , Gene Knock-In Techniques , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Kidney/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Peptide Fragments/genetics , Phenotype , Polycystic Kidney, Autosomal Recessive/metabolism , Receptors, Cell Surface/metabolism
12.
EBioMedicine ; 5: 183-92, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27077126

ABSTRACT

BACKGROUND: The major gene mutated in autosomal dominant polycystic kidney disease was first identified over 20 years ago, yet its function remains poorly understood. We have used a systems-based approach to examine the effects of acquired loss of Pkd1 in adult mouse kidney as it transitions from normal to cystic state. METHODS: We performed transcriptional profiling of a large set of male and female kidneys, along with metabolomics and lipidomics analyses of a subset of male kidneys. We also assessed the effects of a modest diet change on cyst progression in young cystic mice. Fatty acid oxidation and glycolytic rates were measured in five control and mutant pairs of epithelial cells. RESULTS: We find that females have a significantly less severe kidney phenotype and correlate this protection with differences in lipid metabolism. We show that sex is a major determinant of the transcriptional profile of mouse kidneys and that some of this difference is due to genes involved in lipid metabolism. Pkd1 mutant mice have transcriptional profiles consistent with changes in lipid metabolism and distinct metabolite and complex lipid profiles in kidneys. We also show that cells lacking Pkd1 have an intrinsic fatty acid oxidation defect and that manipulation of lipid content of mouse chow modifies cystic disease. INTERPRETATION: Our results suggest PKD could be a disease of altered cellular metabolism.


Subject(s)
Fatty Acids/metabolism , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , TRPP Cation Channels/genetics , Animals , Disease Models, Animal , Female , Humans , Kidney/metabolism , Kidney/pathology , Lipid Metabolism/genetics , Male , Mice , Mice, Transgenic , Oxidation-Reduction , Polycystic Kidney, Autosomal Dominant/physiopathology , TRPP Cation Channels/metabolism
13.
J Am Soc Nephrol ; 26(9): 2081-95, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25952256

ABSTRACT

Polycystic kidney disease (PKD) is one of the most common life-threatening genetic diseases. Jared J. Grantham, M.D., has done more than any other individual to promote PKD research around the world. However, despite decades of investigation there is still no approved therapy for PKD in the United States. In May 2014, the University of Kansas Medical Center hosted a symposium in Kansas City honoring the occasion of Dr. Grantham's retirement and invited all the awardees of the Lillian Jean Kaplan International Prize for Advancement in the Understanding of Polycystic Kidney Disease to participate in a forward-thinking and interactive forum focused on future directions and innovations in PKD research. This article summarizes the contributions of the 12 Kaplan awardees and their vision for the future of PKD research.


Subject(s)
Biomedical Research/trends , Cilia/genetics , Polycystic Kidney, Autosomal Dominant/genetics , Signal Transduction , TRPP Cation Channels/genetics , Animals , Cilia/metabolism , Genes, Modifier , Humans , Kidney Tubules , Mechanistic Target of Rapamycin Complex 1 , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Molecular Targeted Therapy , Multiprotein Complexes/metabolism , Phenotype , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/pathology , Renal Insufficiency/prevention & control , TOR Serine-Threonine Kinases/metabolism , TRPP Cation Channels/metabolism
14.
Article in English | MEDLINE | ID: mdl-25641951

ABSTRACT

The proliferation and diminishing costs of 'omics' approaches have finally opened the doors for small and medium laboratories to enter the 'systems biology era'. This is a welcome evolution that requires a new framework to design, interpret, and validate studies. Here, we highlight some of the challenges, contributions, and prospects of the 'cyst-ems biology' of polycystic kidney disease.


Subject(s)
Kidney/metabolism , Models, Biological , Polycystic Kidney, Autosomal Dominant/metabolism , Proteome/metabolism , TRPP Cation Channels/metabolism , Animals , Computer Simulation , Humans , Systems Biology/methods
15.
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
16.
Mol Biol Cell ; 25(19): 2919-33, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25103236

ABSTRACT

Defects in centrosome and cilium function are associated with phenotypically related syndromes called ciliopathies. Cby1, the mammalian orthologue of the Drosophila Chibby protein, localizes to mature centrioles, is important for ciliogenesis in multiciliated airway epithelia in mice, and antagonizes canonical Wnt signaling via direct regulation of ß-catenin. We report that deletion of the mouse Cby1 gene results in cystic kidneys, a phenotype common to ciliopathies, and that Cby1 facilitates the formation of primary cilia and ciliary recruitment of the Joubert syndrome protein Arl13b. Localization of Cby1 to the distal end of mature centrioles depends on the centriole protein Ofd1. Superresolution microscopy using both three-dimensional SIM and STED reveals that Cby1 localizes to an ∼250-nm ring at the distal end of the mature centriole, in close proximity to Ofd1 and Ahi1, a component of the transition zone between centriole and cilium. The amount of centriole-localized Ahi1, but not Ofd1, is reduced in Cby1(-/-) cells. This suggests that Cby1 is required for efficient recruitment of Ahi1, providing a possible molecular mechanism for the ciliogenesis defect in Cby1(-/-) cells.


Subject(s)
Carrier Proteins/genetics , Centrioles/metabolism , Cilia/genetics , Kidney Diseases, Cystic/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins/metabolism , ADP-Ribosylation Factors/metabolism , Adaptor Proteins, Vesicular Transport , Animals , Carrier Proteins/metabolism , Cell Line , Cilia/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/metabolism , Protein Structure, Tertiary , Proteins/metabolism , Wnt Signaling Pathway , beta Catenin
17.
J Mol Med (Berl) ; 92(10): 1045-56, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24984783

ABSTRACT

Autosomal recessive polycystic kidney disease (ARPKD) results from mutations in the human PKHD1 gene. Both this gene, and its mouse ortholog, Pkhd1, are primarily expressed in renal and biliary ductal structures. The mouse protein product, fibrocystin/polyductin complex (FPC), is a 445-kDa protein encoded by a 67-exon transcript that spans >500 kb of genomic DNA. In the current study, we observed multiple alternatively spliced Pkhd1 transcripts that varied in size and exon composition in embryonic mouse kidney, liver, and placenta samples, as well as among adult mouse pancreas, brain, heart, lung, testes, liver, and kidney. Using reverse transcription PCR and RNASeq, we identified 22 novel Pkhd1 kidney transcripts with unique exon junctions. Various mechanisms of alternative splicing were observed, including exon skipping, use of alternate acceptor/donor splice sites, and inclusion of novel exons. Bioinformatic analyses identified, and exon-trapping minigene experiments validated, consensus binding sites for serine/arginine-rich proteins that modulate alternative splicing. Using site-directed mutagenesis, we examined the functional importance of selected splice enhancers. In addition, we demonstrated that many of the novel transcripts were polysome bound, thus likely translated. Finally, we determined that the human PKHD1 R760H missense variant alters a splice enhancer motif that disrupts exon splicing in vitro and is predicted to truncate the protein. Taken together, these data provide evidence of the complex transcriptional regulation of Pkhd1/PKHD1 and identified motifs that regulate its splicing. Our studies indicate that Pkhd1/PKHD1 transcription is modulated, in part by intragenic factors, suggesting that aberrant PKHD1 splicing represents an unappreciated pathogenic mechanism in ARPKD. Key messages: Multiple mRNA transcripts are generated for Pkhd1 in renal tissues Pkhd1 transcription is modulated by standard splice elements and effectors Mutations in splice motifs may alter splicing to generate nonfunctional peptides.


Subject(s)
Receptors, Cell Surface/genetics , Alternative Splicing , Animals , Exons , Genetic Variation , Humans , Kidney/metabolism , Mice, Inbred DBA , Mutagenesis, Site-Directed , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA , Transcription, Genetic
18.
Cell Rep ; 7(3): 634-44, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24767998

ABSTRACT

Autosomal dominant polycystic kidney disease is a common form of inherited kidney disease that is caused by mutations in two genes, PKD1 (polycystin-1) and PKD2 (polycystin-2). Mice with germline deletion of either gene die in midgestation with a vascular phenotype that includes profound edema. Although an endothelial cell defect has been suspected, the basis of this phenotype remains poorly understood. Here, we demonstrate that edema in Pkd1- and Pkd2-null mice is likely to be caused by defects in lymphatic development. Pkd1 and Pkd2 mutant embryos exhibit reduced lymphatic vessel density and vascular branching along with aberrant migration of early lymphatic endothelial cell precursors. We used cell-based assays to confirm that PKD1- and PKD2-depleted endothelial cells have an intrinsic defect in directional migration that is associated with a failure to establish front-rear polarity. Our studies reveal a role for polycystin signaling in lymphatic development.


Subject(s)
Endothelial Cells/cytology , Lymph Nodes/embryology , Signal Transduction , TRPP Cation Channels/metabolism , Animals , Cell Movement , Cell Polarity , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Lymph Nodes/metabolism , Lymphatic Vessels/embryology , Lymphatic Vessels/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , RNA Interference , RNA, Small Interfering/metabolism , TRPP Cation Channels/antagonists & inhibitors , TRPP Cation Channels/genetics
19.
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
20.
J Am Soc Nephrol ; 25(1): 81-91, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24071006

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is a common cause of renal failure that is due to mutations in two genes, PKD1 and PKD2. Vascular complications, including aneurysms, are a well recognized feature of ADPKD, and a subgroup of families exhibits traits reminiscent of Marfan syndrome (MFS). MFS is caused by mutations in fibrillin-1 (FBN1), which encodes an extracellular matrix protein with homology to latent TGF-ß binding proteins. It was recently demonstrated that fibrillin-1 deficiency is associated with upregulation of TGF-ß signaling. We investigated the overlap between ADPKD and MFS by breeding mice with targeted mutations in Pkd1 and Fbn1. Double heterozygotes displayed an exacerbation of the typical Fbn1 heterozygous aortic phenotype. We show that the basis of this genetic interaction results from further upregulation of TGF-ß signaling caused by Pkd1 haploinsufficiency. In addition, we demonstrate that loss of PKD1 alone is sufficient to induce a heightened responsiveness to TGF-ß. Our data link the interaction of two important diseases to a fundamental signaling pathway.


Subject(s)
Microfilament Proteins/genetics , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , TRPP Cation Channels/genetics , Transforming Growth Factor beta/metabolism , Vascular Diseases/genetics , Vascular Diseases/metabolism , Animals , Disease Models, Animal , Epistasis, Genetic , Female , Fibrillin-1 , Fibrillins , Genetic Association Studies , Haploinsufficiency , Heterozygote , Humans , Male , Marfan Syndrome/etiology , Marfan Syndrome/genetics , Marfan Syndrome/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Microfilament Proteins/deficiency , Mutation , Polycystic Kidney, Autosomal Dominant/complications , Signal Transduction , TRPP Cation Channels/deficiency , Vascular Diseases/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...