Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 70
1.
J Am Soc Nephrol ; 32(11): 2759-2776, 2021 11.
Article En | MEDLINE | ID: mdl-34716241

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD), the most common inherited kidney disease, is regulated by different forms of cell death, including apoptosis and autophagy. However, the role in ADPKD of ferroptosis, a recently discovered form of cell death mediated by iron and lipid metabolism, remains elusive. METHODS: To determine a pathophysiologic role of ferroptosis in ADPKD, we investigated whether the absence of Pkd1 (encoding polycystin-1) affected the expression of key factors involved in the process of ferroptosis, using Western blot and qRT-PCR analysis in Pkd1 mutant renal cells and tissues. We also examined whether treatment with erastin, a ferroptosis inducer, and ferrostain-1, a ferroptosis inhibitor, affected cyst growth in Pkd1 mutant mouse models. RESULTS: We found that kidney cells and tissues lacking Pkd1 exhibit extensive metabolic abnormalities, including reduced expression of the system Xc- amino acid antiporter (critical for import of cystine), of iron exporter (ferroportin), and of GPX4 (a key and negative regulator of ferroptosis). The abnormalities also include increased expression of iron importers (TfR1, DMT1) and HO-1, which in turn result in high iron levels, low GSH and GPX4 activity, increased lipid peroxidation, and propensity to ferroptosis. We further found that erastin increased, and ferrostatin-1 inhibited ferroptotic cell death and proliferation of Pkd1-deficient cells in kidneys from Pkd1 mutant mice. A lipid peroxidation product increased in Pkd1-deficient cells, 4HNE, promoted the proliferation of survived Pkd1 mutant cells via activation of Akt, S6, Stat3, and Rb during the ferroptotic process, contributing to cyst growth. CONCLUSION: These findings indicate that ferroptosis contributes to ADPKD progression and management of ferroptosis may be a novel strategy for ADPKD treatment.


Ferroptosis , Polycystic Kidney, Autosomal Dominant/pathology , Animals , Cell Cycle , Cells, Cultured , Cyclohexylamines/pharmacology , Cyclohexylamines/therapeutic use , Disease Models, Animal , Disease Progression , Epithelial Cells/metabolism , Female , Gene Expression Regulation , Humans , Iron/metabolism , Lipid Peroxidation , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phenylenediamines/pharmacology , Phenylenediamines/therapeutic use , Piperazines/pharmacology , Piperazines/therapeutic use , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/genetics , RNA Interference , Spheroids, Cellular , TRPP Cation Channels/deficiency , TRPP Cation Channels/genetics , Transcriptome
2.
Gene ; 798: 145792, 2021 Sep 25.
Article En | MEDLINE | ID: mdl-34175399

BACKGROUND: Apoptosis is a form of cell death that plays a critical role in the maintenance of tissue homeostasis involving the development and elimination of unwanted cells. Dysregulation of apoptosis appears to be associated in the pathogenesis of many human diseases. Autosomal dominant polycystic kidney disease (ADPKD) is a common monogenetic disease and is mainly caused by mutations in PKD1. Previous studies proved that increased cell death occurred in ADPKD patients and animal models. However, the role of apoptosis in kidney cystogenesis is not clear. METHODS: In current study, due to the high similarities between human and pig, PKD1-deficient (PKD1+/-) pigs and PKD1-knockdown (PKD1KD) pig kidney epithelial cells were used to investigate the mechanisms of apoptosis in driving cystogenesis. RESULTS: In PKD1+/- pigs, increased intrinsic and extrinsic apoptosis were found at ages of 1 month and 3 months, whereas the autophagy and pyroptosis were not altered. Meanwhile, the intrinsic apoptosis was activated along with untouched extrinsic apoptosis in PKD1KD pig kidney cells. Thus, the intrinsic apoptosis played important roles in cystogenesis. CONCLUSIONS: This work provides detail analysis of the roles of different cell death types during cystogenesis in ADPKD pig model. The results suggested a potential new strategy for the diagnosis and treatment of ADPKD by targeting intrinsic apoptosis.


Apoptosis , Polycystic Kidney, Autosomal Dominant/etiology , TRPP Cation Channels/deficiency , Animals , Cell Count , Cell Line , Disease Models, Animal , Gene Knockdown Techniques , Macrophages , Polycystic Kidney, Autosomal Dominant/genetics , Swine , Tumor Necrosis Factor-alpha/metabolism
3.
Biochem Biophys Res Commun ; 561: 73-79, 2021 07 05.
Article En | MEDLINE | ID: mdl-34015761

Metabolic reprogramming is emerging as a key pathological contributor to the progression of autosomal dominant polycystic kidney disease (ADPKD), but the molecular mechanisms underlying dysregulated cellular metabolism remain elusive. Here we report that amino acid biosynthesis is reprogrammed in Pkd2-knockout mouse kidneys via a defective PERK-eIF2ɑ-ATF4 pathway. Transcriptomic analysis revealed that the amino acid biosynthesis pathways such as serine, arginine and cysteine were impaired, and associated critical enzymes were downregulated in Pkd2-knockout mouse kidneys. ATF4 and CHOP, transcription factors downstream of the endoplasmic reticulum (ER) stress sensor PERK, were identified as master regulators of these enzymes' expression. PKD2 deficiency impaired the expression of ATF4 and amino acid synthesis enzymes in RCTEC cells under ER stress. Mechanistically, as an ER-resident protein, PKD2 interacts with TBL2, which functions as an adaptor bridging eIF2ɑ to PERK. PKD2 depletion impaired the recruitment of eIF2ɑ to TBL2, thus impeding activation of the PERK-eIF2ɑ-ATF4 pathway and downstream amino acid biosynthesis. These findings illuminate a molecular mechanism linking the PKD2-mediated PERK-eIF2ɑ-ATF4 pathway and amino acid metabolic reprogramming in ADPKD.


Activating Transcription Factor 4/metabolism , Amino Acids/biosynthesis , Eukaryotic Initiation Factor-2/metabolism , GTP-Binding Proteins/metabolism , Polycystic Kidney, Autosomal Dominant/metabolism , TRPP Cation Channels/deficiency , eIF-2 Kinase/metabolism , Animals , Cells, Cultured , Databases, Genetic , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Humans , Mice , Polycystic Kidney, Autosomal Dominant/pathology , Signal Transduction
4.
J Pathol ; 254(3): 289-302, 2021 07.
Article En | MEDLINE | ID: mdl-33900625

Polycystic liver disease (PLD) is characterized by the growth of numerous biliary cysts and presents in patients with autosomal dominant polycystic kidney disease (ADPKD), causing significant morbidity. Interestingly, deletion of intraflagellar transport-B (IFT-B) complex genes in adult mouse models of ADPKD attenuates the severity of PKD and PLD. Here we examine the role of deletion of an IFT-A gene, Thm1, in PLD of juvenile and adult Pkd2 conditional knockout mice. Perinatal deletion of Thm1 resulted in disorganized and expanded biliary regions, biliary fibrosis, increased serum bile acids, and a shortened primary cilium on cytokeratin 19+ (CK19+) epithelial cells. In contrast, perinatal deletion of Pkd2 caused PLD, with multiple CK19+ epithelial cell-lined cysts, fibrosis, lengthened primary cilia, and increased Notch and ERK signaling. Perinatal deletion of Thm1 in Pkd2 conditional knockout mice increased hepatomegaly, liver necrosis, as well as serum bilirubin and bile acid levels, indicating enhanced liver disease severity. In contrast to effects in the developing liver, deletion of Thm1 alone in adult mice did not cause a biliary phenotype. Combined deletion of Pkd2 and Thm1 caused variable hepatic cystogenesis at 4 months of age, but differences in hepatic cystogenesis between Pkd2- and Pkd2;Thm1 knockout mice were not observed by 6 months of age. Similar to juvenile PLD, Notch and ERK signaling were increased in adult Pkd2 conditional knockout cyst-lining epithelial cells. Taken together, Thm1 is required for biliary tract development, and proper biliary development restricts PLD severity. Unlike IFT-B genes, Thm1 does not markedly attenuate hepatic cystogenesis, suggesting differences in regulation of signaling and cystogenic processes in the liver by IFT-B and -A. Notably, increased Notch signaling in cyst-lining epithelial cells may indicate that aberrant activation of this pathway promotes hepatic cystogenesis, presenting as a novel potential therapeutic target. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Adaptor Proteins, Signal Transducing/deficiency , Biliary Tract/pathology , Polycystic Kidney, Autosomal Dominant/pathology , Animals , Biliary Tract/embryology , Mice , Mice, Knockout , TRPP Cation Channels/deficiency
5.
Semin Cell Dev Biol ; 110: 139-148, 2021 02.
Article En | MEDLINE | ID: mdl-32475690

Polycystic kidney disease (PKD), comprising autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD), is characterized by incessant cyst formation in the kidney and liver. ADPKD and ARPKD represent the leading genetic causes of renal disease in adults and children, respectively. ADPKD is caused by mutations in PKD1 encoding polycystin1 (PC1) and PKD2 encoding polycystin 2 (PC2). PC1/2 are multi-pass transmembrane proteins that form a complex localized in the primary cilium. Predominant ARPKD cases are caused by mutations in polycystic kidney and hepatic disease 1 (PKHD1) gene that encodes the Fibrocystin/Polyductin (FPC) protein, whereas a small subset of cases are caused by mutations in DAZ interacting zinc finger protein 1 like (DZIP1L) gene. FPC is a type I transmembrane protein, localizing to the cilium and basal body, in addition to other compartments, and DZIP1L encodes a transition zone/basal body protein. Apparently, PC1/2 and FPC are signaling molecules, while the mechanism that cilia employ to govern renal tubule morphology and prevent cyst formation is unclear. Nonetheless, recent genetic and biochemical studies offer a glimpse of putative physiological malfunctions and the pathomechanisms underlying both disease entities. In this review, I summarize the results of genetic studies that deduced the function of PC1/2 on cilia and of cilia themselves in cyst formation in ADPKD, and I discuss studies regarding regulation of polycystin biogenesis and cilia trafficking. I also summarize the synergistic genetic interactions between Pkd1 and Pkhd1, and the unique tissue patterning event controlled by FPC, but not PC1. Interestingly, while DZIP1L mutations generate compromised PC1/2 cilia expression, FPC deficiency does not affect PC1/2 biogenesis and ciliary localization, indicating that divergent mechanisms could lead to cyst formation in ARPKD. I conclude by outlining promising areas for future PKD research and highlight rationales for potential therapeutic interventions for PKD treatment.


Adaptor Proteins, Signal Transducing/genetics , Cilia/metabolism , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Recessive/genetics , Receptors, Cell Surface/genetics , TRPP Cation Channels/genetics , Adaptor Proteins, Signal Transducing/deficiency , Adult , Basal Bodies/drug effects , Basal Bodies/metabolism , Basal Bodies/pathology , Child , Cilia/drug effects , Cilia/pathology , Drugs, Chinese Herbal/pharmacology , Flavonoids/pharmacology , Gene Expression , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Mutation , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Polycystic Kidney, Autosomal Recessive/drug therapy , Polycystic Kidney, Autosomal Recessive/metabolism , Polycystic Kidney, Autosomal Recessive/pathology , Receptors, Cell Surface/deficiency , Signal Transduction , TRPP Cation Channels/deficiency
6.
Elife ; 92020 05 04.
Article En | MEDLINE | ID: mdl-32364494

PKD2 (polycystin-2, TRPP1), a TRP polycystin channel, is expressed in endothelial cells (ECs), but its physiological functions in this cell type are unclear. Here, we generated inducible, EC-specific Pkd2 knockout mice to examine vascular functions of PKD2. Data show that a broad range of intravascular flow rates stimulate EC PKD2 channels, producing vasodilation. Flow-mediated PKD2 channel activation leads to calcium influx that activates SK/IK channels and eNOS serine 1176 phosphorylation in ECs. These signaling mechanisms produce arterial hyperpolarization and vasodilation. In contrast, EC PKD2 channels do not contribute to acetylcholine-induced vasodilation, suggesting stimulus-specific function. EC-specific PKD2 knockout elevated blood pressure in mice without altering cardiac function or kidney anatomy. These data demonstrate that flow stimulates PKD2 channels in ECs, leading to SK/IK channel and eNOS activation, hyperpolarization, vasodilation and a reduction in systemic blood pressure. Thus, PKD2 channels are a major component of functional flow sensing in the vasculature.


Arterial Pressure , Endothelial Cells/metabolism , Hypertension/metabolism , Mechanotransduction, Cellular , Mesenteric Arteries/metabolism , TRPP Cation Channels/metabolism , Vasodilation , Animals , Calcium Signaling , Hypertension/genetics , Hypertension/physiopathology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Membrane Potentials , Mesenteric Arteries/physiopathology , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Regional Blood Flow , Small-Conductance Calcium-Activated Potassium Channels/metabolism , TRPP Cation Channels/deficiency , TRPP Cation Channels/genetics
7.
J Am Soc Nephrol ; 31(5): 1035-1049, 2020 05.
Article En | MEDLINE | ID: mdl-32238474

BACKGROUND: The inactivation of the ciliary proteins polycystin 1 or polycystin 2 leads to autosomal dominant polycystic kidney disease (ADPKD). Although signaling by primary cilia and interstitial inflammation both play a critical role in the disease, the reciprocal interactions between immune and tubular cells are not well characterized. The transcription factor STAT3, a component of the cilia proteome that is involved in crosstalk between immune and nonimmune cells in various tissues, has been suggested as a factor fueling ADPKD progression. METHOD: To explore how STAT3 intersects with cilia signaling, renal inflammation, and cyst growth, we used conditional murine models involving postdevelopmental ablation of Pkd1, Stat3, and cilia, as well as cultures of cilia-deficient or STAT3-deficient tubular cell lines. RESULTS: Our findings indicate that, although primary cilia directly modulate STAT3 activation in vitro, the bulk of STAT3 activation in polycystic kidneys occurs through an indirect mechanism in which primary cilia trigger macrophage recruitment to the kidney, which in turn promotes Stat3 activation. Surprisingly, although inactivating Stat3 in Pkd1-deficient tubules slightly reduced cyst burden, it resulted in a massive infiltration of the cystic kidneys by macrophages and T cells, precluding any improvement of kidney function. We also found that Stat3 inactivation led to increased expression of the inflammatory chemokines CCL5 and CXCL10 in polycystic kidneys and cultured tubular cells. CONCLUSIONS: STAT3 appears to repress the expression of proinflammatory cytokines and restrict immune cell infiltration in ADPKD. Our findings suggest that STAT3 is not a critical driver of cyst growth in ADPKD but rather plays a major role in the crosstalk between immune and tubular cells that shapes disease expression.


Kidney Tubules/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , STAT3 Transcription Factor/physiology , Aged, 80 and over , Animals , Cells, Cultured , Chemokine CCL5/metabolism , Chemokine CXCL10/metabolism , Cilia/metabolism , Dogs , Humans , Inflammation , Kidney Tubules/pathology , Macrophages/physiology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred C57BL , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/immunology , Polycystic Kidney, Autosomal Dominant/metabolism , Specific Pathogen-Free Organisms , T-Lymphocytes/physiology , TRPP Cation Channels/deficiency , TRPP Cation Channels/metabolism
8.
J Mol Med (Berl) ; 98(5): 659-671, 2020 05.
Article En | MEDLINE | ID: mdl-32185407

Polycystic kidney disease (PKD) leads to continuous decline of renal function by growth of renal cysts. Enhanced proliferation and transepithelial chloride secretion through cystic fibrosis transmembrane conductance regulator (CFTR) and Ca2+-activated TMEM16A Cl- channels is thought to cause an increase in cyst volume. Recent work shows the pro-proliferative role of the Ca2+ activated Cl- channel TMEM16A (anoctamin 1), and demonstrates the essential contribution of TMEM16A to CFTR-dependent Cl- secretion. The present data demonstrate an increase in intracellular Ca2+ ([Ca2+]i) signals and Cl- secretion by TMEM16A, in renal collecting duct principle cells from dog (MDCK) and mouse (M1) as well as primary tubular epithelial cells from PKD1-/- knockout mice. M1 organoids proliferated, increased expression of TMEM16A, and secreted Cl- upon knockdown of endogenous polycystin 1 or 2 (PKD1,2), by retroviral transfection with shPKD1 and shPKD2, respectively. Knockdown of PKD1 or PKD2 increased basal intracellular Ca2+ levels and enhanced purinergic Ca2+ release from endoplasmic reticulum. In contrast, ryanodine receptors were found not to be expressed in mouse renal epithelial cells and caffeine had no effects on [Ca2+]i. Ca2+ signals, proliferation, and Cl- secretion were largely reduced by knockdown or blockade of TMEM16A. TMEM16A may be therefore important for enhanced Ca2+ release from IP3-sensitive Ca2+ stores in polycystic kidney disease. KEY MESSAGES: • ADPKD leads to continuous decline of renal function by growth of renal cysts. • Knockdown of PKD1 or PKD2 increases TMEM16A expression. • TMEM16A enhanced intracellular Ca2+ signals, Cl- secretion, and proliferation. • TMEM16A contributes to cyst growth in ADPKD.


Anoctamin-1/genetics , Anoctamin-1/metabolism , Calcium Signaling , Calcium/metabolism , Disease Susceptibility , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Action Potentials , Adenosine Triphosphate/metabolism , Animals , Cell Line , Cell Proliferation , Chlorides/metabolism , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Intracellular Space/metabolism , Ion Transport , Mice , Mice, Knockout , Polycystic Kidney Diseases/pathology , Protein Kinase C/deficiency , TRPP Cation Channels/deficiency
9.
Sci Rep ; 10(1): 72, 2020 01 09.
Article En | MEDLINE | ID: mdl-31919453

Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenic cause of end-stage renal failure in humans and results from germline mutations in PKD1 or PKD2. Despite the recent approval of tolvaptan, safer and more effective alternative drugs are clearly needed to slow disease progression. As a first step in drug discovery, we conducted an unbiased chemical screen on zebrafish pkd2 mutant embryos using two publicly available compound libraries (Spectrum, PKIS) totalling 2,367 compounds to identify novel treatments for ADPKD. Using dorsal tail curvature as the assay readout, three major chemical classes (steroids, coumarins, flavonoids) were identified from the Spectrum library as the most promising candidates to be tested on human PKD1 cystic cells. Amongst these were an androgen, 5α-androstane 3,17-dione, detected as the strongest enhancer of the pkd2 phenotype but whose effect was found to be independent of the canonical androgen receptor pathway. From the PKIS library, we identified several ALK5 kinase inhibitors as strong suppressors of the pkd2 tail phenotype and in vitro cyst expansion. In summary, our results identify ALK5 and non-canonical androgen receptors as potential therapeutic targets for further evaluation in drug development for ADPKD.


Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , TRPP Cation Channels/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified/metabolism , Apoptosis/drug effects , Dogs , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , High-Throughput Screening Assays , Humans , Madin Darby Canine Kidney Cells , Phenotype , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptors, Androgen/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , TRPP Cation Channels/deficiency , TRPP Cation Channels/metabolism , Zebrafish , Zebrafish Proteins/deficiency , Zebrafish Proteins/metabolism
10.
Methods Cell Biol ; 154: 109-120, 2019.
Article En | MEDLINE | ID: mdl-31493813

Oligonucleotides are small molecules 8-50 nucleotides in length that bind via Watson-Crick base pairing to enhance or repress the expression of target RNA. The use of oligonucleotides to manipulate gene expression in the kidney could be a valuable tool to further understand kidney pathophysiology and can serve as an important complement to genetic studies. This chapter serves as a primer on the use of oligonucleotides in the kidney. We provide an overview of the various ways that oligonucleotides can manipulate gene expression. In addition, we describe the advancements in the development of oligonucleotides for laboratory and clinical use. Finally, instruction is provided on the design and implementation of oligonucleotides for in vitro and in vivo laboratory studies.


Genetic Therapy/methods , Kidney/metabolism , MicroRNAs/genetics , Oligonucleotides, Antisense/genetics , Polycystic Kidney Diseases/therapy , TRPP Cation Channels/genetics , Animals , Cell Line , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Humans , Kidney/pathology , Mice , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/metabolism , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology , TRPP Cation Channels/deficiency
11.
Am J Physiol Lung Cell Mol Physiol ; 317(4): L466-L474, 2019 10 01.
Article En | MEDLINE | ID: mdl-31411061

The transient receptor potential polycystin-2 (TRPP2) is encoded by the Pkd2 gene, and mutation of this gene can cause autosomal dominant polycystic kidney disease (ADPKD). Some patients with ADPKD experience extrarenal manifestations, including radiologic and clinical bronchiectasis. We hypothesized that TRPP2 may regulate airway smooth muscle (ASM) tension. Thus, we used smooth muscle-Pkd2 conditional knockout (Pkd2SM-CKO) mice to investigate whether TRPP2 regulated ASM tension and whether TRPP2 deficiency contributed to bronchiectasis associated with ADPKD. Compared with wild-type mice, Pkd2SM-CKO mice breathed more shallowly and faster, and their cross-sectional area ratio of bronchi to accompanying pulmonary arteries was higher, suggesting that TRPP2 may regulate ASM tension and contribute to the occurrence of bronchiectasis in ADPKD. In a bioassay examining isolated tracheal ring tension, no significant difference was found for high-potassium-induced depolarization of the ASM between the two groups, indicating that TRPP2 does not regulate depolarization-induced ASM contraction. By contrast, carbachol-induced contraction of the ASM derived from Pkd2SM-CKO mice was significantly reduced compared with that in wild-type mice. In addition, relaxation of the carbachol-precontracted ASM by isoprenaline, a ß-adrenergic receptor agonist that acts through the cAMP/adenylyl cyclase pathway, was also significantly attenuated in Pkd2SM-CKO mice compared with that in wild-type mice. Thus, TRPP2 deficiency suppressed both contraction and relaxation of the ASM. These results provide a potential target for regulating ASM tension and for developing therapeutic alternatives for some ADPKD complications of the respiratory system or for independent respiratory disease, especially bronchiectasis.


Bronchi/metabolism , Bronchiectasis/genetics , Muscle, Smooth/metabolism , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics , Animals , Bronchi/drug effects , Bronchi/physiopathology , Bronchiectasis/metabolism , Bronchiectasis/physiopathology , Bronchodilator Agents/antagonists & inhibitors , Bronchodilator Agents/pharmacology , Calcium/metabolism , Carbachol/pharmacology , Disease Models, Animal , Gene Expression Regulation , Isometric Contraction/drug effects , Isometric Contraction/physiology , Isoproterenol/antagonists & inhibitors , Isoproterenol/pharmacology , Male , Mice , Mice, Knockout , Miotics/pharmacology , Muscle Tonus/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiopathology , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/physiopathology , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Respiration/drug effects , Signal Transduction , TRPP Cation Channels/deficiency , Trachea/drug effects , Trachea/metabolism , Trachea/physiopathology
12.
Sci Adv ; 5(6): eaaw3593, 2019 06.
Article En | MEDLINE | ID: mdl-31183407

Positive transcription elongation factor b (P-TEFb) functions as a central regulator of transcription elongation. Activation of P-TEFb occurs through its dissociation from the transcriptionally inactive P-TEFb/HEXIM1/7SK snRNP complex. However, the mechanisms of signal-regulated P-TEFb activation and its roles in human diseases remain largely unknown. Here, we demonstrate that cAMP-PKA signaling disrupts the inactive P-TEFb/HEXIM1/7SK snRNP complex by PKA-mediated phosphorylation of HEXIM1 at serine-158. The cAMP pathway plays central roles in the development of autosomal dominant polycystic kidney disease (ADPKD), and we show that P-TEFb is hyperactivated in mouse and human ADPKD kidneys. Genetic activation of P-TEFb promotes cyst formation in a zebrafish ADPKD model, while pharmacological inhibition of P-TEFb attenuates cyst development by suppressing the pathological gene expression program in ADPKD mice. Our study therefore elucidates a mechanism by which P-TEFb activation by cAMP-PKA signaling promotes cystogenesis in ADPKD.


Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Positive Transcriptional Elongation Factor B/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/chemistry , Cysts/metabolism , Cysts/pathology , Disease Models, Animal , Flavonoids/pharmacology , Flavonoids/therapeutic use , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Mice , Mice, Knockout , Phosphorylation , Piperidines/pharmacology , Piperidines/therapeutic use , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/metabolism , Positive Transcriptional Elongation Factor B/antagonists & inhibitors , Positive Transcriptional Elongation Factor B/genetics , Protein Binding , RNA-Binding Proteins/metabolism , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/metabolism , Signal Transduction , TRPP Cation Channels/deficiency , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism , Transcription Factors/metabolism , Zebrafish/metabolism
13.
Circulation ; 140(11): 921-936, 2019 09 10.
Article En | MEDLINE | ID: mdl-31220931

BACKGROUND: Polycystin-1 (PC1) is a transmembrane protein originally identified in autosomal dominant polycystic kidney disease where it regulates the calcium-permeant cation channel polycystin-2. Autosomal dominant polycystic kidney disease patients develop renal failure, hypertension, left ventricular hypertrophy, and diastolic dysfunction, among other cardiovascular disorders. These individuals harbor PC1 loss-of-function mutations in their cardiomyocytes, but the functional consequences are unknown. PC1 is ubiquitously expressed, and its experimental ablation in cardiomyocyte-specific knockout mice reduces contractile function. Here, we set out to determine the pathophysiological role of PC1 in cardiomyocytes. METHODS: Wild-type and cardiomyocyte-specific PC1 knockout mice were analyzed by echocardiography. Excitation-contraction coupling was assessed in isolated cardiomyocytes and human embryonic stem cell-derived cardiomyocytes, and functional consequences were explored in heterologous expression systems. Protein-protein interactions were analyzed biochemically and by means of ab initio calculations. RESULTS: PC1 ablation reduced action potential duration in cardiomyocytes, decreased Ca2+ transients, and myocyte contractility. PC1-deficient cardiomyocytes manifested a reduction in sarcoendoplasmic reticulum Ca2+ stores attributable to a reduced action potential duration and sarcoendoplasmic reticulum Ca2+ ATPase (SERCA) activity. An increase in outward K+ currents decreased action potential duration in cardiomyocytes lacking PC1. Overexpression of full-length PC1 in HEK293 cells significantly reduced the current density of heterologously expressed Kv4.3, Kv1.5 and Kv2.1 potassium channels. PC1 C terminus inhibited Kv4.3 currents to the same degree as full-length PC1. Additionally, PC1 coimmunoprecipitated with Kv4.3, and a modeled PC1 C-terminal structure suggested the existence of 2 docking sites for PC1 within the N terminus of Kv4.3, supporting a physical interaction. Finally, a naturally occurring human mutant PC1R4228X manifested no suppressive effects on Kv4.3 channel activity. CONCLUSIONS: Our findings uncover a role for PC1 in regulating multiple Kv channels, governing membrane repolarization and alterations in SERCA activity that reduce cardiomyocyte contractility.


Action Potentials/physiology , Myocytes, Cardiac/metabolism , Potassium Channels, Voltage-Gated/metabolism , TRPP Cation Channels/deficiency , Animals , Humans , Mice , Mice, Knockout , Mice, Transgenic , TRPP Cation Channels/genetics
14.
Cell Physiol Biochem ; 52(5): 1061-1074, 2019.
Article En | MEDLINE | ID: mdl-30977988

BACKGROUND/AIMS: Autosomal dominant polycystic kidney disease (ADPKD) is characterized by progressive cyst formation and growth, leading to end-stage renal disease. A higher kidney volume is predictive of a more accelerated decline in renal function. This study aimed to examine the effects of caffeine, a phosphodiesterase inhibitor, on the progression of cystic kidney disease in a mouse model orthologous to human disease (Pkd1cond/cond:Nestincre). METHODS: Caffeine was administered to male cystic (CyCaf) and noncystic (NCCaf) mice (Pkd1cond/cond) from conception and at the postweaning period through 12 weeks of life (3 mg/d), while control animals consumed water (CyCtrl and NCCtrl). Renal ultrasonography was performed at 10 weeks of life to calculate total kidney volume and cystic index. At the end of the protocol, blood and urine samples were collected for biochemical analysis, and animals were euthanized. Kidneys were harvested to obtain renal tissue for determinations of adenosine 3´5´-cyclic monophosphate (cAMP) by an enzymatic immunoassay kit and phosphorylated extracellular signal-regulated kinase (p-ERK) by Western blotting. Renal fibrosis (picrosirius staining), renal cell proliferation (ki-67 immunohistochemistry) and apoptotic rates (TUNEL analysis) were also determined. RESULTS: At 12 weeks, CyCaf mice exhibited higher serum urea nitrogen, renal cystic index, total kidney volume, kidney cell proliferation, apoptosis and fibrosis compared with CyCtrl mice. Serum cystatin C was significantly higher in CyCaf than in NCCaf and NCCtrl mice. CyCaf mice had higher total kidney weight than all other groups but not higher heart and liver weight. The levels of cAMP and p-ERK did not differ among the groups. CONCLUSION: Caffeine consumption from conception through 12 weeks led to increased cystic index and total kidney volume and worsened renal function in Pkd1-deficient cystic mice, suggesting that high consumption of caffeine may contribute to a faster progression of renal disease in ADPKD.


Caffeine/adverse effects , Kidney/metabolism , Polycystic Kidney Diseases , TRPP Cation Channels/deficiency , Animals , Caffeine/pharmacology , Cyclic AMP/genetics , Cyclic AMP/metabolism , Disease Models, Animal , Kidney/pathology , Male , Mice , Mice, Knockout , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology
15.
Circulation ; 139(20): 2342-2357, 2019 05 14.
Article En | MEDLINE | ID: mdl-30818997

BACKGROUND: The primary cilium is a singular cellular structure that extends from the surface of many cell types and plays crucial roles in vertebrate development, including that of the heart. Whereas ciliated cells have been described in developing heart, a role for primary cilia in adult heart has not been reported. This, coupled with the fact that mutations in genes coding for multiple ciliary proteins underlie polycystic kidney disease, a disorder with numerous cardiovascular manifestations, prompted us to identify cells in adult heart harboring a primary cilium and to determine whether primary cilia play a role in disease-related remodeling. METHODS: Histological analysis of cardiac tissues from C57BL/6 mouse embryos, neonatal mice, and adult mice was performed to evaluate for primary cilia. Three injury models (apical resection, ischemia/reperfusion, and myocardial infarction) were used to identify the location and cell type of ciliated cells with the use of antibodies specific for cilia (acetylated tubulin, γ-tubulin, polycystin [PC] 1, PC2, and KIF3A), fibroblasts (vimentin, α-smooth muscle actin, and fibroblast-specific protein-1), and cardiomyocytes (α-actinin and troponin I). A similar approach was used to assess for primary cilia in infarcted human myocardial tissue. We studied mice silenced exclusively in myofibroblasts for PC1 and evaluated the role of PC1 in fibrogenesis in adult rat fibroblasts and myofibroblasts. RESULTS: We identified primary cilia in mouse, rat, and human heart, specifically and exclusively in cardiac fibroblasts. Ciliated fibroblasts are enriched in areas of myocardial injury. Transforming growth factor ß-1 signaling and SMAD3 activation were impaired in fibroblasts depleted of the primary cilium. Extracellular matrix protein levels and contractile function were also impaired. In vivo, depletion of PC1 in activated fibroblasts after myocardial infarction impaired the remodeling response. CONCLUSIONS: Fibroblasts in the neonatal and adult heart harbor a primary cilium. This organelle and its requisite signaling protein, PC1, are required for critical elements of fibrogenesis, including transforming growth factor ß-1-SMAD3 activation, production of extracellular matrix proteins, and cell contractility. Together, these findings point to a pivotal role of this organelle, and PC1, in disease-related pathological cardiac remodeling and suggest that some of the cardiovascular manifestations of autosomal dominant polycystic kidney disease derive directly from myocardium-autonomous abnormalities.


Fibroblasts/ultrastructure , Myocardium/pathology , Polycystic Kidney, Autosomal Dominant/pathology , 3T3 Cells/ultrastructure , Animals , Animals, Newborn , Atrial Remodeling , Cilia , Fetal Heart/cytology , Fibrosis , Heart Injuries/pathology , Humans , Kinesins/deficiency , Kinesins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Polycystic Kidney, Autosomal Dominant/genetics , Rats , Signal Transduction , Smad3 Protein/physiology , TRPP Cation Channels/deficiency , TRPP Cation Channels/physiology , Transforming Growth Factor beta1/physiology , Ventricular Remodeling
16.
Elife ; 72018 12 04.
Article En | MEDLINE | ID: mdl-30511640

Systemic blood pressure is determined, in part, by arterial smooth muscle cells (myocytes). Several Transient Receptor Potential (TRP) channels are proposed to be expressed in arterial myocytes, but it is unclear if these proteins control physiological blood pressure and contribute to hypertension in vivo. We generated the first inducible, smooth muscle-specific knockout mice for a TRP channel, namely for PKD2 (TRPP1), to investigate arterial myocyte and blood pressure regulation by this protein. Using this model, we show that intravascular pressure and α1-adrenoceptors activate PKD2 channels in arterial myocytes of different systemic organs. PKD2 channel activation in arterial myocytes leads to an inward Na+ current, membrane depolarization and vasoconstriction. Inducible, smooth muscle cell-specific PKD2 knockout lowers both physiological blood pressure and hypertension and prevents pathological arterial remodeling during hypertension. Thus, arterial myocyte PKD2 controls systemic blood pressure and targeting this TRP channel reduces high blood pressure.


Arteries/metabolism , Hypertension/genetics , Myocytes, Smooth Muscle/metabolism , Receptors, Adrenergic, alpha-1/genetics , Sodium/metabolism , TRPP Cation Channels/genetics , Animals , Arteries/physiopathology , Blood Pressure/physiology , Cations, Monovalent , Gene Expression Regulation , Hindlimb/blood supply , Hindlimb/cytology , Hypertension/metabolism , Hypertension/physiopathology , Ion Transport , Membrane Potentials/physiology , Mice , Mice, Knockout , Myocytes, Smooth Muscle/pathology , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction , TRPP Cation Channels/deficiency , Vasoconstriction/physiology
17.
Biomed Pharmacother ; 108: 1123-1134, 2018 Dec.
Article En | MEDLINE | ID: mdl-30372813

Mutations in the PKD1 or PKD2 genes are the cause of autosomal dominant polycystic kidney disease (ADPKD). The encoded proteins localize within the cell membrane and primary cilia and are proposed to be involved in mechanotransduction. Therefore, we evaluate shear stress dependent signaling in renal epithelial cells and the relevance for ADPKD. Using RNA sequencing and pathway analysis, we compared gene expression of in vitro shear stress treated Pkd1-/- renal epithelial cells and in vivo pre-cystic Pkd1del models. We show that shear stress alters the same signaling pathways in Pkd1-/- renal epithelial cells and Pkd1wt controls. However, expression of a number of genes was slightly more induced by shear stress in Pkd1-/- cells, suggesting that Pkd1 has the function to restrain shear regulated signaling instead of being a mechano-sensing activator. We also compared altered gene expression in Pkd1-/- cells during shear with in vivo transcriptome data of kidneys from Pkd1del mice at three early pre-cystic time-points. This revealed overlap of a limited number of differentially expressed genes. However, the overlap between cells and mice is much higher when looking at pathways and molecular processes, largely due to altered expression of paralogous genes. Several of the altered pathways in the in vitro and in vivo Pkd1del models are known to be implicated in ADPKD pathways, including PI3K-AKT, MAPK, Hippo, calcium, Wnt, and TGF-ß signaling. We hypothesize that increased activation of selected genes in renal epithelial cells early upon Pkd1 gene disruption may disturb the balance in signaling and may contribute to cyst formation.


Kidney Tubules, Proximal/pathology , Polycystic Kidney Diseases/genetics , Signal Transduction/genetics , Stress, Mechanical , TRPP Cation Channels/deficiency , Animals , Cilia/metabolism , Epithelial Cells/metabolism , Gene Deletion , Gene Expression Profiling , Male , Mice , Organ Size , Polycystic Kidney Diseases/pathology , TRPP Cation Channels/metabolism , Transcription, Genetic
18.
Am J Physiol Renal Physiol ; 315(6): F1855-F1868, 2018 12 01.
Article En | MEDLINE | ID: mdl-30280600

Research into metabolic reprogramming in cancer has become commonplace, yet this area of research has only recently come of age in nephrology. In light of the parallels between cancer and autosomal dominant polycystic kidney disease (ADPKD), the latter is currently being studied as a metabolic disease. In clear cell renal cell carcinoma (RCC), which is now considered a metabolic disease, we and others have shown derangements in the enzyme arginosuccinate synthase 1 (ASS1), resulting in RCC cells becoming auxotrophic for arginine and leading to a new therapeutic paradigm involving reducing extracellular arginine. Based on our earlier finding that glutamine pathways are reprogrammed in ARPKD, and given the connection between arginine and glutamine synthetic pathways via citrulline, we investigated the possibility of arginine reprogramming in ADPKD. We now show that, in a remarkable parallel to RCC, ASS1 expression is reduced in murine and human ADPKD, and arginine depletion results in a dose-dependent compensatory increase in ASS1 levels as well as decreased cystogenesis in vitro and ex vivo with minimal toxicity to normal cells. Nontargeted metabolomics analysis of mouse kidney cell lines grown in arginine-deficient versus arginine-replete media suggests arginine-dependent alterations in the glutamine and proline pathways. Thus, depletion of this conditionally essential amino acid by dietary or pharmacological means, such as with arginine-degrading enzymes, may be a novel treatment for this disease.


Arginine/metabolism , Cell Proliferation , Energy Metabolism , Kidney/metabolism , Polycystic Kidney, Autosomal Dominant/metabolism , Animals , Arginine/deficiency , Arginine/pharmacology , Argininosuccinate Synthase/genetics , Argininosuccinate Synthase/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Energy Metabolism/drug effects , Female , Genetic Predisposition to Disease , Humans , Kidney/drug effects , Kidney/pathology , Male , Metabolomics/methods , Mice, Knockout , Phenotype , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/pathology , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Signal Transduction , TRPP Cation Channels/deficiency , TRPP Cation Channels/genetics
19.
J Am Soc Nephrol ; 29(10): 2471-2481, 2018 10.
Article En | MEDLINE | ID: mdl-30209078

BACKGROUND: In patients with autosomal dominant polycystic kidney disease (ADPKD), most of whom have a mutation in PKD1 or PKD2, abnormally large numbers of macrophages accumulate around kidney cysts and promote their growth. Research by us and others has suggested that monocyte chemoattractant protein-1 (Mcp1) may be a signal for macrophage-mediated cyst growth. METHODS: To define the role of Mcp1 and macrophages in promoting cyst growth, we used mice with inducible knockout of Pkd1 alone (single knockout) or knockout of both Pkd1 and Mcp1 (double knockout) in the murine renal tubule. Levels of Mcp1 RNA expression were measured in single-knockout mice and controls. RESULTS: In single-knockout mice, upregulation of Mcp1 precedes macrophage infiltration. Macrophages accumulating around nascent cysts (0-2 weeks after induction) are initially proinflammatory and induce tubular cell injury with morphologic flattening, oxidative DNA damage, and proliferation-independent cystic dilation. At 2-6 weeks after induction, macrophages switch to an alternative activation phenotype and promote further cyst growth because of an additional three-fold increase in tubular cell proliferative rates. In double-knockout mice, there is a marked reduction in Mcp1 expression and macrophage numbers, resulting in less initial tubular cell injury, slower cyst growth, and improved renal function. Treatment of single-knockout mice with an inhibitor to the Mcp1 receptor Ccr2 partially reproduced the morphologic and functional improvement seen with Mcp1 knockout. CONCLUSIONS: Mcp1 is upregulated after knockout of Pkd1 and promotes macrophage accumulation and cyst growth via both proliferation-independent and proliferation-dependent mechanisms in this orthologous mouse model of ADPKD.


Chemokine CCL2/genetics , Chemokine CCL2/physiology , Macrophages/physiology , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/pathology , Animals , Chemokine CCL2/deficiency , DNA Damage , Disease Models, Animal , Humans , Kidney Tubules/pathology , Kidney Tubules/physiopathology , Macrophage Activation/drug effects , Macrophage Activation/genetics , Macrophage Activation/physiology , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Knockout , Polycystic Kidney, Autosomal Dominant/physiopathology , Pyrrolidines/pharmacology , Reactive Oxygen Species/metabolism , Receptors, CCR2/antagonists & inhibitors , TRPP Cation Channels/deficiency , TRPP Cation Channels/genetics , Up-Regulation
20.
Am J Physiol Renal Physiol ; 315(3): F537-F546, 2018 09 01.
Article En | MEDLINE | ID: mdl-29767557

The PKD1 gene encodes polycystin-1 (PC1), a mechanosensor triggering intracellular responses upon urinary flow sensing in kidney tubular cells. Mutations in PKD1 lead to autosomal dominant polycystic kidney disease (ADPKD). The involvement of PC1 in renal electrolyte handling remains unknown since renal electrolyte physiology in ADPKD patients has only been characterized in cystic ADPKD. We thus studied the renal electrolyte handling in inducible kidney-specific Pkd1 knockout (iKsp- Pkd1-/-) mice manifesting a precystic phenotype. Serum and urinary electrolyte determinations indicated that iKsp- Pkd1-/- mice display reduced serum levels of magnesium (Mg2+), calcium (Ca2+), sodium (Na+), and phosphate (Pi) compared with control ( Pkd1+/+) mice and renal Mg2+, Ca2+, and Pi wasting. In agreement with these electrolyte disturbances, downregulation of key genes for electrolyte reabsorption in the thick ascending limb of Henle's loop (TA;, Cldn16, Kcnj1, and Slc12a1), distal convoluted tubule (DCT; Trpm6 and Slc12a3) and connecting tubule (CNT; Calb1, Slc8a1, and Atp2b4) was observed in kidneys of iKsp- Pkd1-/- mice compared with controls. Similarly, decreased renal gene expression of markers for TAL ( Umod) and DCT ( Pvalb) was observed in iKsp- Pkd1-/- mice. Conversely, mRNA expression levels in kidney of genes encoding solute and water transporters in the proximal tubule ( Abcg2 and Slc34a1) and collecting duct ( Aqp2, Scnn1a, and Scnn1b) remained comparable between control and iKsp- Pkd1-/- mice, although a water reabsorption defect was observed in iKsp- Pkd1-/- mice. In conclusion, our data indicate that PC1 is involved in renal Mg2+, Ca2+, and water handling and its dysfunction, resulting in a systemic electrolyte imbalance characterized by low serum electrolyte concentrations.


Body Water/metabolism , Electrolytes/metabolism , Kidney/metabolism , Polycystic Kidney, Autosomal Dominant/metabolism , TRPP Cation Channels/deficiency , Water-Electrolyte Balance , Animals , Calcium/metabolism , Disease Models, Animal , Electrolytes/blood , Electrolytes/urine , Gene Expression Regulation , Intestinal Absorption , Kidney/physiopathology , Magnesium/metabolism , Male , Mice, Knockout , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/physiopathology , Renal Reabsorption , TRPP Cation Channels/genetics , Water-Electrolyte Balance/genetics
...