Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.246
Filter
Add more filters

Publication year range
1.
FASEB J ; 37(7): e23008, 2023 07.
Article in English | MEDLINE | ID: mdl-37318790

ABSTRACT

Autosomal recessive polycystic kidney disease (ARPKD) is caused primarily by mutations in PKHD1, encoding fibrocystin (FPC), but Pkhd1 mutant mice failed to reproduce the human phenotype. In contrast, the renal lesion in congenital polycystic kidney (cpk) mice, with a mutation in Cys1 and cystin protein loss, closely phenocopies ARPKD. Although the nonhomologous mutation diminished the translational relevance of the cpk model, recent identification of patients with CYS1 mutations and ARPKD prompted the investigations described herein. We examined cystin and FPC expression in mouse models (cpk, rescued-cpk (r-cpk), Pkhd1 mutants) and mouse cortical collecting duct (CCD) cell lines (wild type (wt), cpk). We found that cystin deficiency caused FPC loss in both cpk kidneys and CCD cells. FPC levels increased in r-cpk kidneys and siRNA of Cys1 in wt cells reduced FPC. However, FPC deficiency in Pkhd1 mutants did not affect cystin levels. Cystin deficiency and associated FPC loss impacted the architecture of the primary cilium, but not ciliogenesis. No reduction in Pkhd1 mRNA levels in cpk kidneys and CCD cells suggested posttranslational FPC loss. Studies of cellular protein degradation systems suggested selective autophagy as a mechanism. In support of the previously described function of FPC in E3 ubiquitin ligase complexes, we demonstrated reduced polyubiquitination and elevated levels of functional epithelial sodium channel in cpk cells. Therefore, our studies expand the function of cystin in mice to include inhibition of Myc expression via interaction with necdin and maintenance of FPC as functional component of the NEDD4 E3 ligase complexes. Loss of FPC from E3 ligases may alter the cellular proteome, contributing to cystogenesis through multiple, yet to be defined, mechanisms.


Subject(s)
Polycystic Kidney, Autosomal Recessive , Humans , Mice , Animals , Polycystic Kidney, Autosomal Recessive/genetics , Polycystic Kidney, Autosomal Recessive/metabolism , Polycystic Kidney, Autosomal Recessive/pathology , Proteome/metabolism , Receptors, Cell Surface/metabolism , Kidney/metabolism , Transcription Factors/metabolism , Epithelial Cells/metabolism
2.
Am J Physiol Renal Physiol ; 317(3): F632-F637, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31313950

ABSTRACT

Flow-related bending of cilia results in Ca2+ influx through a polycystin-1 (Pkd1) and polycystin-2 (Pkd2) complex, both of which are members of the transient receptor potential (TRP) family (TRPP1 and TRPP2, respectively). Deletion of this complex as well as cilia result in polycystic kidney disease. The Ca2+ influx pathway has been previously characterized in immortalized collecting duct cells without cilia and found to be a 23-pS channel that was a multimere of TRPP2 and TRPV4. The purpose of the present study was to determine if this TRPP2 and TRPV4 multimere exists in vivo. Apical channel activity was measured using the patch-clamp technique from isolated split-open cortical collecting ducts from adult conditional knockout mice with (Ift88flox/flox) or without (Ift88-/-) cilia. Single tubules were isolated for measurements of mRNA for Pkd1, Pkd2, Trpv4, and epithelial Na+ channel subunits. The predominant channel activity from Ift88flox/flox mice was from epithelial Na+ channel [5-pS Na+-selective channels with long mean open times (475.7 ± 83.26 ms) and open probability > 0.2]. With the loss of cilia, the predominant conductance was a 23-pS nonselective cation channel (reversal potential near 0) with a short mean open time (72 ± 17 ms), open probability < 0.08, and a characteristic flickery opening. Loss of cilia increased mRNA levels for Pkd2 and Trpv4 from single isolated cortical collecting ducts. In conclusion, 23-pS channels exist in vivo, and activity of this channel is elevated with loss of cilia, consistent with previous finding of an elevated-unregulated Ca2+-permeable pathway at the apical membrane of collecting duct cells that lack cilia.


Subject(s)
Cilia/metabolism , Kidney Tubules, Collecting/metabolism , Polycystic Kidney Diseases/metabolism , TRPP Cation Channels/metabolism , TRPV Cation Channels/metabolism , Animals , Calcium Signaling , Cilia/pathology , Disease Models, Animal , Female , Kidney Tubules, Collecting/pathology , Male , Membrane Potentials , Mice, Knockout , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/pathology , TRPP Cation Channels/genetics , TRPV Cation Channels/genetics , Time Factors , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Up-Regulation
3.
Am J Physiol Renal Physiol ; 316(3): F414-F425, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30566001

ABSTRACT

Deficiency in polycystin 1 triggers specific changes in energy metabolism. To determine whether defects in other human cystoproteins have similar effects, we studied extracellular acidification and glucose metabolism in human embryonic kidney (HEK-293) cell lines with polycystic kidney and hepatic disease 1 ( PKHD1) and polycystic kidney disease (PKD) 2 ( PKD2) truncating defects along multiple sites of truncating mutations found in patients with autosomal recessive and dominant PKDs. While neither the PKHD1 or PKD2 gene mutations nor their position enhanced cell proliferation rate in our cell line models, truncating mutations in these genes progressively increased overall extracellular acidification over time ( P < 0.001 for PKHD1 and PKD2 mutations). PKHD1 mutations increased nonglycolytic acidification rate (1.19 vs. 1.03, P = 0.002), consistent with an increase in tricarboxylic acid cycle activity or breakdown of intracellular glycogen. In addition, they increased basal and ATP-linked oxygen consumption rates [7.59 vs. 5.42 ( P = 0.015) and 4.55 vs. 2.98 ( P = 0.004)]. The PKHD1 and PKD2 mutations also altered mitochondrial morphology, resembling the effects of polycystin 1 deficiency. Together, these data suggest that defects in major PKD genes trigger changes in mitochondrial energy metabolism. After validation in in vivo models, these initial observations would indicate potential benefits of targeting energy metabolism in the treatment of PKDs.


Subject(s)
Energy Metabolism/genetics , Glucose/metabolism , Protein Kinases/genetics , Receptors, Cell Surface/genetics , Cell Proliferation/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing , HEK293 Cells , Humans , Mutation , Protein Kinase D2 , Protein Kinases/metabolism , Receptors, Cell Surface/metabolism
4.
FASEB J ; 30(1): 370-9, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26391272

ABSTRACT

Activation of the intrarenal renin angiotensin system (RAS) is believed to play an important role in the development of hypertension and cystogenesis in autosomal dominant polycystic kidney disease (ADPKD). Results of clinical studies testing RAS inhibitors in slowing the progression of cystic disease in ADPKD are inconclusive, and we hypothesized that current RAS inhibitors do not adequately suppress intrarenal RAS. For this study, we compared a novel Gen 2 antisense oligonucleotide (ASO) that inhibits angiotensinogen (Agt) synthesis to lisinopril in adult conditional Pkd1 systemic-knockout mice, a model of ADPKD. Six weeks after Pkd1 global gene knockout, the mice were treated with Agt-ASO (66 mg/kg/wk), lisinopril (100 mg/kg/d), PBS (control), or scrambled ASO (66 mg/kg/wk) for 10 wk, followed by tissue collection. Agt ASO resulted in significant reduction in plasma, liver, and kidney Agt, and increased kidney renin compared with control treatments. Kidneys from Agt-ASO-treated mice were not as enlarged and showed reduced cystic volume compared with lisinopril or control treatments. Blood pressure was better controlled with lisinopril than with Agt-ASO. Agt-ASO suppressed cell proliferation in both cystic and noncystic cells compared with lisinopril and control treatments. However, Agt-ASO did not reduce cell proliferation in liver, which indicates that Agt-ASO targets cell signaling pathways that specifically suppresses cystogenesis in the kidney. These data suggest that Agt-ASO effectively attenuates intrarenal RAS and therefore can be a novel and effective agent for treating ADPKD.


Subject(s)
Angiotensinogen/metabolism , Polycystic Kidney Diseases/metabolism , TRPP Cation Channels/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensinogen/biosynthesis , Animals , Female , Kidney/drug effects , Kidney/metabolism , Lisinopril/pharmacology , Male , Mice , Mice, Inbred C57BL , Polycystic Kidney Diseases/genetics , TRPP Cation Channels/genetics
5.
Br J Sports Med ; 51(7): 607-611, 2017 Apr.
Article in English | MEDLINE | ID: mdl-26755678

ABSTRACT

OBJECTIVE: The primary aims of this retrospective study were to describe the burden of injury presenting to the medical team and the changes in injury profile over 10 years (2003-2012) at The Championships, Wimbledon. Secondary aims included description of gender difference in rates, distribution and pathology of injuries. DESIGN: Retrospective observational cohort of player injury presentations over 10 years (2003-2012) at The Championships, Wimbledon. RESULTS: The overall rate of presentation of injury for all players over the 10-year period was 20.7 per 1000 sets played. Injury rates were lower for male players (17.7 injuries per 1000 sets played) than female players (23.4 injuries per 1000 sets played). There was variability in the numbers of injuries reported by men and women players over the 10-year period. CONCLUSIONS: The rates of presentation of injury at this Grand Slam tennis tournament varied between male and female players, and between years. More robust systems of data collection are required in professional tennis to enable more sophisticated injury data analysis between sexes, years and different playing surfaces.


Subject(s)
Athletic Injuries/epidemiology , Tennis/injuries , Athletes , Female , Humans , Incidence , Male , Retrospective Studies
6.
J Biol Chem ; 290(11): 6890-902, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25616662

ABSTRACT

Microtubule-based centrioles in the centrosome mediate accurate bipolar cell division, spindle orientation, and primary cilia formation. Cellular checkpoints ensure that the centrioles duplicate only once in every cell cycle and achieve precise dimensions, dysregulation of which results in genetic instability and neuro- and ciliopathies. The normal cellular level of centrosomal protein 4.1-associated protein (CPAP), achieved by its degradation at mitosis, is considered as one of the major mechanisms that limits centriole growth at a predetermined length. Here we show that CPAP levels and centriole elongation are regulated by centrobin. Exogenous expression of centrobin causes abnormal elongation of centrioles due to massive accumulation of CPAP in the cell. Conversely, CPAP was undetectable in centrobin-depleted cells, suggesting that it undergoes degradation in the absence of centrobin. Only the reintroduction of full-length centrobin, but not its mutant form that lacks the CPAP binding site, could restore cellular CPAP levels in centrobin-depleted cells, indicating that persistence of CPAP requires its interaction with centrobin. Interestingly, inhibition of the proteasome in centrobin-depleted cells restored the cellular and centriolar CPAP expression, suggesting its ubiquitination and proteasome-mediated degradation when centrobin is absent. Intriguingly, however, centrobin-overexpressing cells also showed proteasome-independent accumulation of ubiquitinated CPAP and abnormal, ubiquitin-positive, elongated centrioles. Overall, our results show that centrobin interacts with ubiquitinated CPAP and prevents its degradation for normal centriole elongation function. Therefore, it appears that loss of centrobin expression destabilizes CPAP and triggers its degradation to restrict the centriole length during biogenesis.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/metabolism , Microtubule-Associated Proteins/metabolism , Cell Cycle Proteins/analysis , Cell Cycle Proteins/genetics , Cell Line , Centrioles/ultrastructure , Gene Deletion , Humans , Microtubule-Associated Proteins/analysis , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitination , Up-Regulation
7.
Physiology (Bethesda) ; 30(3): 195-207, 2015 May.
Article in English | MEDLINE | ID: mdl-25933820

ABSTRACT

Autosomal-dominant polycystic kidney disease (ADPKD) is the most prevalent inherited renal disease, characterized by multiple cysts that can eventually lead to kidney failure. Studies investigating the role of primary cilia and polycystins have significantly advanced our understanding of the pathogenesis of PKD. This review will present clinical and basic aspects of ADPKD, review current concepts of PKD pathogenesis, evaluate potential therapeutic targets, and highlight challenges for future clinical studies.


Subject(s)
Kidney , Mutation , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/therapy , TRPP Cation Channels/genetics , Animals , Cilia , Genetic Predisposition to Disease , Genetic Testing , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Molecular Targeted Therapy , Phenotype , Polycystic Kidney, Autosomal Dominant/diagnosis , Polycystic Kidney, Autosomal Dominant/epidemiology , Polycystic Kidney, Autosomal Dominant/metabolism , Predictive Value of Tests , Risk Factors , Signal Transduction , Treatment Outcome
8.
Clin Endocrinol (Oxf) ; 85(3): 386-92, 2016 09.
Article in English | MEDLINE | ID: mdl-27175553

ABSTRACT

CONTEXT: In observational studies, low serum 25-hydroxyvitamin D (25-OHD) concentration is associated with an increased risk of type 2 diabetes mellitus (DM). Increasing serum 25-OHD may have beneficial effects on insulin resistance or beta-cell function. Cross-sectional studies utilizing suboptimal methods for assessment of insulin sensitivity and serum 25-OHD concentration provide conflicting results. OBJECTIVE: This study examined the relationship between serum 25-OHD concentration and insulin resistance in healthy overweight individuals at increased risk of cardiovascular disease, using optimal assessment techniques. METHODS: A total of 92 subjects (mean age 56·0, SD 6·0 years), who were healthy but overweight (mean body mass index 30·9, SD 2·3 kg/m(2) ), underwent assessments of insulin sensitivity (two-step euglycaemic hyperinsulinaemic clamp, HOMA2-IR), beta-cell function (HOMA2%B), serum 25-OHD concentration and body composition (DEXA). RESULTS: Mean total 25-OHD concentration was 32·2, range 21·8-46·6 nmol/l. No association was demonstrated between serum 25-OHD concentration and insulin resistance. CONCLUSIONS: In this study using optimal assessment techniques to measure 25-OHD concentration, insulin sensitivity and body composition, there was no association between serum 25-OHD concentration and insulin resistance in healthy, overweight individuals at high risk of developing cardiovascular disease. This study suggests the documented inverse association between serum 25-OHD concentration and risk of type 2 DM is not mediated by a relationship between serum 25-OHD concentration and insulin resistance.


Subject(s)
Vitamin D/analogs & derivatives , Cardiovascular Diseases , Cross-Sectional Studies , Diabetes Mellitus, Type 2 , Humans , Insulin Resistance , Middle Aged , Overweight , Vitamin D/blood
9.
Proteomics ; 15(23-24): 4051-63, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26364553

ABSTRACT

Proteomic studies including marine mammals are rare, largely due to the lack of fully sequenced genomes. This has hampered the application of these techniques toward biomarker discovery efforts for monitoring of health and disease in these animals. We conducted a pilot label-free LC-MS/MS study to profile and compare the cerebrospinal fluid from California sea lions with domoic acid toxicosis (DAT) and without DAT. Across 11 samples, a total of 206 proteins were identified (FDR<0.1) using a composite mammalian database. Several peptide identifications were validated using stable isotope labeled peptides. Comparison of spectral counts revealed seven proteins that were elevated in the cerebrospinal fluid from sea lions with DAT: complement C3, complement factor B, dickkopf-3, malate dehydrogenase 1, neuron cell adhesion molecule 1, gelsolin, and neuronal cell adhesion molecule. Immunoblot analysis found reelin to be depressed in the cerebrospinal fluid from California sea lions with DAT. Mice administered domoic acid also had lower hippocampal reelin protein levels suggesting that domoic acid depresses reelin similar to kainic acid. In summary, proteomic analysis of cerebrospinal fluid in marine mammals is a useful tool to characterize the underlying molecular pathology of neurodegenerative disease. All MS data have been deposited in the ProteomeXchange with identifier PXD002105 (http://proteomecentral.proteomexchange.org/dataset/PXD002105).


Subject(s)
Kainic Acid/analogs & derivatives , Neurodegenerative Diseases/metabolism , Proteomics , Sea Lions/metabolism , Animals , Kainic Acid/cerebrospinal fluid , Kainic Acid/metabolism , Reelin Protein , Sea Lions/genetics
10.
Am J Physiol Renal Physiol ; 309(6): F492-8, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26155843

ABSTRACT

Since polycystic kidney disease (PKD) was first noted over 30 years ago to have neoplastic parallels, there has been a resurgent interest in elucidating neoplasia-relevant pathways in PKD. Taking a nontargeted metabolomics approach in the B6(Cg)-Cys1(cpk/)J (cpk) mouse model of recessive PKD, we have now characterized metabolic reprogramming in these tissues, leading to a glutamine-dependent TCA cycle shunt toward total 2-hydroxyglutarate (2-HG) production in cpk compared with B6 wild-type kidney tissue. After confirmation of increased 2-HG expression in immortalized collecting duct cpk cells as well as in human autosomal recessive PKD tissue using targeted analysis, we show that the increase in 2-HG is likely due to glutamine-sourced α-ketoglutarate. In addition, cpk cells require exogenous glutamine for growth such that inhibition of glutaminase-1 decreases cell viability as well as proliferation. This study is a demonstration of the striking parallels between recessive PKD and cancer metabolism. Our data, once confirmed in other PKD models, suggest that future therapeutic approaches targeting this pathway, such as using glutaminase inhibitors, have the potential to open novel treatment options for renal cystic disease.


Subject(s)
Creatine Kinase/genetics , Glutamine/metabolism , Glutarates/metabolism , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Animals , Cells, Cultured , DNA Mutational Analysis , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Glutaminase/antagonists & inhibitors , Humans , Infant , Infant, Newborn , Male , Metabolomics , Mice , Models, Genetic
11.
Am J Physiol Renal Physiol ; 309(1): F79-87, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25904703

ABSTRACT

In polycystic kidney disease (PKD), the rate of cyst formation and disease progression is highly variable. The lack of predictability in disease progression may be due to additional environmental factors or pathophysiological processes called "third hits." Diabetes is a growing epidemic, and recent studies suggest that PKD patients may be at an increased risk for this disease. We sought to determine if hyperglycemia enhances the initiation and rate of cystogenesis. Tamoxifen was administered to adult Ift88 conditional floxed allele mice to induce cilia loss in the presence of Cre. Subsequent administration of streptozotocin resulted in equivalent hyperglycemia in cilia(+) and cilia(-) mice. Hyperglycemia with loss of cilia increased the rate of cyst formation and cell proliferation. Structural and functional alterations in the kidney, including focal glomerular foot process effacement, interstitial inflammation, formation of primitive renal tubules, polyuria, and increased proteinuria, were also observed in hyperglycemic cilia(-) mice. Gene array analysis indicated enhanced Wnt and epithelial-to-mesenchymal transition signaling in the kidney of hyperglycemic cilia(-) mice. These data show that hyperglycemia, in the absence of cilia, results in renal structural and functional damage and accelerates cystogenesis, suggesting that diabetes is a risk factor in the progression of PKD.


Subject(s)
Hyperglycemia/complications , Kidney/pathology , Polycystic Kidney Diseases/etiology , Animals , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Hemodynamics , Hyperglycemia/pathology , Hyperglycemia/physiopathology , Kidney Function Tests , Male , Mice, Knockout , Polycystic Kidney Diseases/pathology , Random Allocation , Wnt Proteins/metabolism
13.
J Am Soc Nephrol ; 25(6): 1187-97, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24511141

ABSTRACT

Domoic acid (DA), an excitatory amino acid produced by diatoms belonging to the genus Pseudo-nitzschia, is a glutamate analog responsible for the neurologic condition referred to as amnesic shellfish poisoning. To date, the renal effects of DA have been underappreciated, although renal filtration is the primary route of systemic elimination and the kidney expresses ionotropic glutamate receptors. To characterize the renal effects of DA, we administered either a neurotoxic dose of DA or doses below the recognized limit of toxicity to adult Sv128/Black Swiss mice. DA preferentially accumulated in the kidney and elicited marked renal vascular and tubular damage consistent with acute tubular necrosis, apoptosis, and renal tubular cell desquamation, with toxic vacuolization and mitochondrial swelling as hallmarks of the cellular damage. Doses≥0.1 mg/kg DA elevated the renal injury biomarkers kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin, and doses≥0.005 mg/kg induced the early response genes c-fos and junb. Coadministration of DA with the broad spectrum excitatory amino acid antagonist kynurenic acid inhibited induction of c-fos, junb, and neutrophil gelatinase-associated lipocalin. These findings suggest that the kidney may be susceptible to excitotoxic agonists, and renal effects should be considered when examining glutamate receptor activation. Additionally, these results indicate that DA is a potent nephrotoxicant, and potential renal toxicity may require consideration when determining safe levels for human exposure.


Subject(s)
Kainic Acid/analogs & derivatives , Marine Toxins/toxicity , Neuromuscular Depolarizing Agents/toxicity , Neuromuscular Junction/drug effects , Animals , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Kainic Acid/pharmacokinetics , Kainic Acid/toxicity , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Marine Toxins/pharmacokinetics , Mice, Inbred Strains , Microscopy, Electron, Transmission , Mitochondrial Swelling/drug effects , Myocardium/metabolism , Myocardium/pathology , Neuromuscular Depolarizing Agents/pharmacokinetics , Neuromuscular Junction/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Kainic Acid/genetics , Receptors, Kainic Acid/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Vacuoles/pathology , Vacuoles/ultrastructure , GluK2 Kainate Receptor
14.
Am J Physiol Cell Physiol ; 307(6): C554-60, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25055824

ABSTRACT

Epidermal growth factor (EGF) is linked to the pathogenesis of polycystic kidney disease (PKD). We explored signaling pathways activated by EGF in orpk cilia (-) collecting duct cell line derived from a mouse model of PKD (hypomorph of the Tg737/Ift88 gene) with severely stunted cilia, and in a control orpk cilia (+) cell line with normal cilia. RT-PCR demonstrated mRNAs for EGF receptor subunits ErbB1, ErbB2, ErbB3, ErbB4, and mRNAs for Na(+)/H(+) exchangers (NHE), NHE-1, NHE-2, NHE-3, NHE-4, and NHE-5 in both cell lines. EGF stimulated proton efflux in both cell lines. This effect was significantly attenuated by MIA, 5-(n-methyl-N-isobutyl) amiloride, a selective inhibitor of NHE-1 and NHE-2, and orpk cilia (-) cells were more sensitive to MIA than control cells (P < 0.01). EGF significantly induced extracellular signal-regulated kinase (ERK) phosphorylation in both cilia (+) and cilia (-) cells (63.3 and 123.6%, respectively), but the effect was more pronounced in orpk cilia (-) cells (P < 0.01). MIA significantly attenuated EGF-induced ERK phosphorylation only in orpk cilia (-) cells (P < 0.01). EGF increased proliferation of orpk cilia (+) cells and orpk cilia (-) cells, respectively, and MIA at 1-5 µM attenuated EGF-induced proliferation in orpk cilia (-) cells without affecting proliferation of orpk cilia (+) cells. EGF-induced proliferation of both cell lines was significantly decreased by the EGFR tyrosine kinase inhibitor AG1478 and MEK inhibitor PD98059. These results suggest that EGF exerts mitogenic effects in the orpk cilia (-) cells via activation of growth-associated amiloride-sensitive NHEs and ERK.


Subject(s)
Cell Proliferation , Epidermal Growth Factor/metabolism , Kidney Tubules, Collecting/enzymology , Polycystic Kidney Diseases/enzymology , Sodium-Hydrogen Exchangers/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line , Cell Proliferation/drug effects , Cilia/enzymology , Cilia/pathology , Disease Models, Animal , Enzyme Activation , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Isoenzymes , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/pathology , Mice , Mice, Transgenic , Phosphorylation , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/pathology , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Signal Transduction , Sodium-Hydrogen Exchangers/genetics , Transfection , Tumor Suppressor Proteins/genetics
15.
Am J Physiol Renal Physiol ; 307(5): F551-9, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24899059

ABSTRACT

Polycystic kidney disease (PKD) is a common genetic disorder leading to cyst formation in the kidneys and other organs that ultimately results in kidney failure and death. Currently, there is no therapy for slowing down or stopping the progression of PKD. In this study, we identified the disintegrin metalloenzyme 17 (ADAM17) as a key regulator of cell proliferation in kidney tissues of conditional knockout Ift88(-/-) mice and collecting duct epithelial cells from Ift88°(rpk) mice, animal models of autosomal recessive polycystic kidney disease (ARPKD). Using Western blotting, an enzyme activity assay, and a growth factor-shedding assay in the presence or absence of the specific ADAM17 inhibitor TMI-005, we show that increased expression and activation of ADAM17 in the cystic kidney and in collecting duct epithelial cells originating from the Ift88°(rpk) mice (designated as PKD cells) lead to constitutive shedding of several growth factors, including heparin-binding EGF-like growth factor (HB-EGF), amphiregulin, and transforming growth factor-α (TGF-α). Increased growth factor shedding induces activation of the EGFR/MAPK/ERK pathway and maintains higher cell proliferation rate in PKD cells compared with control cells. PKD cells also displayed increased lactate formation and extracellular acidification indicative of aerobic glycolysis (Warburg effect), which was blocked by ADAM17 inhibition. We propose that ADAM17 is a key promoter of cellular proliferation in PKD cells by activating the EGFR/ERK axis and a proproliferative glycolytic phenotype.


Subject(s)
ADAM Proteins/physiology , Cell Proliferation/physiology , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/physiology , Glycolysis/physiology , Kidney Tubules, Collecting/pathology , Polycystic Kidney Diseases/physiopathology , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/drug effects , ADAM17 Protein , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/physiology , ErbB Receptors/physiology , Female , Heparin-binding EGF-like Growth Factor/physiology , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/physiopathology , Male , Mice , Mice, Knockout , Morpholines/pharmacology , Phenotype , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/pathology , Transforming Growth Factor alpha/physiology , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
16.
Am J Physiol Renal Physiol ; 307(5): F560-70, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24920756

ABSTRACT

Nearly all patients with tuberous sclerosis complex (TSC) develop renal angiomyolipomas, although the tumor cell of origin is unknown. We observed decreased renal angiomyolipoma development in patients with TSC2- polycystic kidney disease 1 deletion syndrome and hypertension that were treated from an early age with angiotensin-converting enzyme inhibitors or angiotensin receptor blockers compared with patients who did not receive this therapy. TSC-associated renal angiomyolipomas expressed ANG II type 1 receptors, platelet-derived growth factor receptor-ß, desmin, α-smooth muscle actin, and VEGF receptor 2 but did not express the adipocyte marker S100 or the endothelial marker CD31. Sera of TSC patients exhibited increased vascular mural cell-secreted peptides, such as VEGF-A, VEGF-D, soluble VEGF receptor 2, and collagen type IV. These findings suggest that angiomyolipomas may arise from renal pericytes. ANG II treatment of angiomyolipoma cells in vitro resulted in an exaggerated intracellular Ca(2+) response and increased proliferation, which were blocked by the ANG II type 2 receptor antagonist valsartan. Blockade of ANG II signaling may have preventative therapeutic potential for angiomyolipomas.


Subject(s)
Angiomyolipoma/drug therapy , Angiomyolipoma/pathology , Angiotensin Receptor Antagonists/therapeutic use , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Pericytes/pathology , Tuberous Sclerosis/complications , Angiomyolipoma/physiopathology , Angiotensin II/physiology , Angiotensin Receptor Antagonists/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Calcium/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Humans , In Vitro Techniques , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/physiopathology , Receptor, Angiotensin, Type 1/physiology , Renin-Angiotensin System/physiology , Signal Transduction/physiology , Tetrazoles/pharmacology , Tetrazoles/therapeutic use , Tuberous Sclerosis/pathology , Tuberous Sclerosis/physiopathology , Valine/analogs & derivatives , Valine/pharmacology , Valine/therapeutic use , Valsartan
17.
Am J Physiol Lung Cell Mol Physiol ; 306(2): L162-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24213915

ABSTRACT

The mechanisms for the development of bronchiectasis and airway hyperreactivity have not been fully elucidated. Although genetic, acquired diseases and environmental influences may play a role, it is also possible that motile cilia can influence this disease process. We hypothesized that deletion of a key intraflagellar transport molecule, IFT88, in mature mice causes loss of cilia, resulting in airway remodeling. Airway cilia were deleted by knockout of IFT88, and airway remodeling and pulmonary function were evaluated. In IFT88(-) mice there was a substantial loss of airway cilia on respiratory epithelium. Three months after the deletion of cilia, there was clear evidence for bronchial remodeling that was not associated with inflammation or apparent defects in mucus clearance. There was evidence for airway epithelial cell hypertrophy and hyperplasia. IFT88(-) mice exhibited increased airway reactivity to a methacholine challenge and decreased ciliary beat frequency in the few remaining cells that possessed cilia. With deletion of respiratory cilia there was a marked increase in the number of club cells as seen by scanning electron microscopy. We suggest that airway remodeling may be exacerbated by the presence of club cells, since these cells are involved in airway repair. Club cells may be prevented from differentiating into respiratory epithelial cells because of a lack of IFT88 protein that is necessary to form a single nonmotile cilium. This monocilium is a prerequisite for these progenitor cells to transition into respiratory epithelial cells. In conclusion, motile cilia may play an important role in controlling airway structure and function.


Subject(s)
Bronchial Hyperreactivity/pathology , Bronchiectasis/pathology , Cilia/pathology , Cilia/physiology , Ciliary Motility Disorders/pathology , Animals , Bronchial Hyperreactivity/physiopathology , Bronchiectasis/physiopathology , Bronchoconstrictor Agents/pharmacology , Ciliary Motility Disorders/physiopathology , Disease Models, Animal , Methacholine Chloride/pharmacology , Mice , Mice, Knockout , Mucociliary Clearance/physiology , Respiratory Mucosa/drug effects , Respiratory Mucosa/pathology , Respiratory Mucosa/physiopathology , Tumor Suppressor Proteins/genetics
18.
Phys Rev Lett ; 112(10): 105003, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24679301

ABSTRACT

Indirect drive experiments at the National Ignition Facility are designed to achieve fusion by imploding a fuel capsule with x rays from a laser-driven hohlraum. Previous experiments have been unable to determine whether a deficit in measured ablator implosion velocity relative to simulations is due to inadequate models of the hohlraum or ablator physics. ViewFactor experiments allow for the first time a direct measure of the x-ray drive from the capsule point of view. The experiments show a 15%-25% deficit relative to simulations and thus explain nearly all of the disagreement with the velocity data. In addition, the data from this open geometry provide much greater constraints on a predictive model of laser-driven hohlraum performance than the nominal ignition target.

19.
Scand J Med Sci Sports ; 24(3): 477-90, 2014 Jun.
Article in English | MEDLINE | ID: mdl-23710994

ABSTRACT

Recently, cherries and cherry products have received growing attention within the literature with regard to their application in both exercise and clinical paradigms. Reported to be high in anti-inflammatory and anti-oxidative capacity, cherries and their constituents are proposed to provide a similar but natural alternative akin to over-the-counter non-steroidal anti-inflammatory drugs (NSAIDs) or analgesics. Within exercise paradigms, concern has been raised with regard to the use of products, which inhibit such inflammatory or oxidative actions, because of the possibility of the blunting of physiological training adaptations. Despite this, numerous scenarios exist both within exercise and clinical populations where a goal of optimal recovery time is more important than physiological adaptation. This review critically evaluates and discusses the use of cherries as a supplementation strategy to enhance recovery of muscle function, inhibit exercise-induced inflammation, oxidative stress, and pain primarily; furthermore, the potential application of cherries to clinical populations is discussed.


Subject(s)
Exercise/physiology , Inflammation/physiopathology , Muscle, Skeletal/physiology , Oxidative Stress/physiology , Prunus , Diet , Humans , Inflammation/prevention & control , Musculoskeletal Pain/physiopathology , Musculoskeletal Pain/prevention & control , Recovery of Function/physiology
20.
PLoS Genet ; 7(4): e1001361, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21490950

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is characterized by formation of renal cysts that destroy the kidney. Mutations in PKD1 and PKD2, encoding polycystins-1 and -2, cause ADPKD. Polycystins are thought to function in primary cilia, but it is not well understood how these and other proteins are targeted to cilia. Here, we provide the first genetic and biochemical link between polycystins and the exocyst, a highly-conserved eight-protein membrane trafficking complex. We show that knockdown of exocyst component Sec10 yields cellular phenotypes associated with ADPKD, including loss of flow-generated calcium increases, hyperproliferation, and abnormal activation of MAPK. Sec10 knockdown in zebrafish phenocopies many aspects of polycystin-2 knockdown-including curly tail up, left-right patterning defects, glomerular expansion, and MAPK activation-suggesting that the exocyst is required for pkd2 function in vivo. We observe a synergistic genetic interaction between zebrafish sec10 and pkd2 for many of these cilia-related phenotypes. Importantly, we demonstrate a biochemical interaction between Sec10 and the ciliary proteins polycystin-2, IFT88, and IFT20 and co-localization of the exocyst and polycystin-2 at the primary cilium. Our work supports a model in which the exocyst is required for the ciliary localization of polycystin-2, thus allowing for polycystin-2 function in cellular processes.


Subject(s)
Phenotype , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , TRPP Cation Channels/metabolism , Vesicular Transport Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Carrier Proteins/metabolism , Cell Line , Cilia/genetics , Cilia/metabolism , Dogs , Enzyme Activation/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , HEK293 Cells , Humans , Kidney/embryology , Kidney/pathology , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Polycystic Kidney Diseases/pathology , Protein Binding , TRPP Cation Channels/deficiency , Tumor Suppressor Proteins/metabolism , Vesicular Transport Proteins/genetics , Zebrafish , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL