Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Int J Med Sci ; 16(1): 180-188, 2019.
Article in English | MEDLINE | ID: mdl-30662341

ABSTRACT

Aim: Autosomal dominant polycystic kidney disease is one of the most common genetic renal diseases. Cyclooxygenase plays an important role in epithelial cell proliferation and may contribute to the mechanisms underlying cyst formation. The aim of the present study was to evaluate the role of cyclooxygenase inhibition in the cyst progression in polycystic kidney disease. Method: Pkd2WS25/- mice, a murine model which harbors a compound cis-heterozygous mutation of the Pkd2 gene were used. Cyclooxygenase expression was assessed in both human and murine kidney specimens. Pkd2WS25/- mice were treated with Sulindac (a nonselective cyclooxygenase inhibitor) or vehicle for 8 months starting at three weeks age, and then renal cyst burden was assessed by kidney weight and volume. Results: Cyclooxygenase-2 expression was up-regulated compared to control kidneys as shown by RNase protection in human polycystic kidneys and immunoblot in mouse Pkd2WS25/- kidneys. Cyclooxygenase-2 expression was up-regulated in the renal interstitium as well as focal areas of the cystic epithelium (p<0.05). Basal Cyclooxygenase-1 levels were unchanged in both immunohistochemistry and real-time PCR. Administration of Sulindac to Pkd2WS25/- mice and to control mice for 8 months resulted in reduced kidney weights and volume in cystic mice. Renal function and electrolytes were not significantly different between groups. Conclusion: Thus treatment of a murine model of polycystic kidney disease with Sulindac results in decreased kidney cyst burden. These findings provide additional implications for the use of Cyclooxygenase inhibition as treatment to slow the progression of cyst burden in patients with polycystic kidney disease.


Subject(s)
Cyclooxygenase Inhibitors/therapeutic use , Polycystic Kidney, Autosomal Dominant/drug therapy , Sulindac/therapeutic use , Animals , Cell Proliferation/drug effects , Cysts/metabolism , Cysts/physiopathology , Dinoprostone/biosynthesis , Disease Models, Animal , Disease Progression , Glomerular Filtration Rate/drug effects , Humans , Mice , Molecular Targeted Therapy , Mutation , Prostaglandin-E Synthases/biosynthesis , Prostaglandin-Endoperoxide Synthases/biosynthesis , Prostaglandins/biosynthesis , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism
2.
J Exp Clin Cancer Res ; 37(1): 89, 2018 Apr 27.
Article in English | MEDLINE | ID: mdl-29699590

ABSTRACT

BACKGROUND: Dysregulation of transforming growth factor ß (TGF-ß) signaling and hypoxic microenvironment have respectively been reported to be involved in disease progression in malignancies of prostate. Emerging evidence indicates that downregulation of TGFBR2, a pivotal regulator of TGF-ß signaling, may contribute to carcinogenesis and progression of prostate cancer (PCa). However, the biological function and regulatory mechanism of TGFBR2 in PCa remain poorly understood. In this study, we propose to investigate the crosstalk of hypoxia and TGF-ß signaling and provide insight into the molecular mechanism underlying the regulatory pathways in PCa. METHODS: Prostate cancer cell lines were cultured in hypoxia or normoxia to evaluate the effect of hypoxia on TGFBR2 expression. Methylation specific polymerase chain reaction (MSP) and demethylation agents was used to evaluate the methylation regulation of TGFBR2 promoter. Besides, silencing of EZH2 via specific siRNAs or chemical inhibitor was used to validate the regulatory effect of EZH2 on TGFBR2. Moreover, we conducted PCR, western blot, and luciferase assays which studied the relationship of miR-93 and TGFBR2 in PCa cell lines and specimens. We also detected the impacts of hypoxia on EZH2 and miR-93, and further examined the tumorigenic functions of miR-93 on proliferation and epithelial-mesenchymal transition via a series of experiments. RESULTS: TGFBR2 expression was attenuated under hypoxia. Hypoxia-induced EZH2 promoted H3K27me3 which caused TGFBR2 promoter hypermethylation and contributed to its epigenetic silencing in PCa. Besides, miR-93 was significantly upregulated in PCa tissues and cell lines, and negatively correlated with the expression of TGFBR2. Ectopic expression of miR-93 promoted cell proliferation, migration and invasion in PCa, and its expression could also be induced by hypoxia. In addition, TGFBR2 was identified as a bona fide target of miR-93. CONCLUSIONS: Our findings elucidate diverse hypoxia-regulated pathways including EZH2-mediated hypermethylation and miR-93-induced silencing contribute to attenuation of TGFBR2 expression and promote cancer progression in prostate cancer.


Subject(s)
Prostatic Neoplasms/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Cell Hypoxia , Cell Line, Tumor , Disease Progression , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Transfection
3.
JCI Insight ; 3(5)2018 03 08.
Article in English | MEDLINE | ID: mdl-29515026

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) can be caused by mutations in the PKD1 or PKD2 genes. The PKD1 gene product is a Wnt cell-surface receptor. We previously showed that a lack of the PKD2 gene product, PC2, increases ß-catenin signaling in mouse embryonic fibroblasts, kidney renal epithelia, and isolated renal collecting duct cells. However, it remains unclear whether ß-catenin signaling plays a role in polycystic kidney disease phenotypes or if a Wnt inhibitor can halt cyst formation in ADPKD disease models. Here, using genetic and pharmacologic approaches, we demonstrated that the elevated ß-catenin signaling caused by PC2 deficiency contributes significantly to disease phenotypes in a mouse ortholog of human ADPKD. Pharmacologically inhibiting ß-catenin stability or the production of mature Wnt protein, or genetically reducing the expression of Ctnnb1 (which encodes ß-catenin), suppressed the formation of renal cysts, improved renal function, and extended survival in ADPKD mice. Our study clearly demonstrates the importance of ß-catenin signaling in disease phenotypes associated with Pkd2 mutation. It also describes the effects of two Wnt inhibitors, XAV939 and LGK974, on various Wnt signaling targets as a potential therapeutic modality for ADPKD, for which there is currently no effective therapy.


Subject(s)
Heterocyclic Compounds, 3-Ring/administration & dosage , Polycystic Kidney, Autosomal Dominant/drug therapy , Pyrazines/administration & dosage , Pyridines/administration & dosage , Wnt Signaling Pathway/drug effects , beta Catenin/antagonists & inhibitors , Animals , Disease Models, Animal , Female , Humans , Injections, Intraperitoneal , Kidney/drug effects , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/mortality , Polycystic Kidney, Autosomal Dominant/pathology , Random Allocation , Survival Analysis , TRPP Cation Channels/genetics , Treatment Outcome , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
4.
Sci Rep ; 7(1): 11493, 2017 09 13.
Article in English | MEDLINE | ID: mdl-28904368

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in PKD1 or PKD2 which encodes polycystin-1 (PC1) and polycystin-2, respectively. PC1 was previously shown to slow cell proliferation and inhibit apoptosis but the underlying mechanisms remain elusive or controversial. Here we showed in cultured mammalian cells and Pkd1 knockout mouse kidney epithelial cells that PC1 and its truncation mutant comprising the last five transmembrane segments and the intracellular C-terminus (PC1-5TMC) down-regulate the phosphorylation of protein kinase R (PKR) and its substrate eukaryotic translation initiation factor 2 alpha (eIF2α). PKR is known to be activated by interferons and dsRNAs, inhibits protein synthesis and induces apoptosis. By co-immunoprecipitation experiments we found that PC1 truncation mutants associate with PKR, or with PKR and its activator PACT. Further experiments showed that PC1 and PC1-5TMC reduce phosphorylation of eIF2α through inhibiting PKR phosphorylation. Our TUNEL experiments using tunicamycin, an apoptosis inducer, and GADD34, an inhibitor of eIF2α phosphorylation, demonstrated that PC1-5TMC inhibits apoptosis of HEK293T cells in a PKR-eIF2α-dependent manner, with concurrent up- and down-regulation of Bcl-2 and Bax, respectively, revealed by Western blotting. Involvement of PC1-regulated eIF2α phosphorylation and a PKR-eIF2α pathway in cell apoptosis may be an important part of the mechanism underlying ADPKD pathogenesis.


Subject(s)
Apoptosis , Eukaryotic Initiation Factor-2/metabolism , Signal Transduction , TRPP Cation Channels/metabolism , eIF-2 Kinase/metabolism , Animals , Apoptosis/genetics , Eukaryotic Initiation Factor-2/antagonists & inhibitors , HEK293 Cells , HeLa Cells , Humans , Kidney/metabolism , Mice , Phosphorylation , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Protein Binding , Protein Interaction Domains and Motifs , eIF-2 Kinase/genetics
5.
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
6.
Curr Gene Ther ; 17(1): 43-49, 2017.
Article in English | MEDLINE | ID: mdl-28494735

ABSTRACT

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease in the clinic. The predominant clinical manifestation is bilateral and progressive cysts formation in the kidneys, impairs normal renal parenchyma, and ultimately leads to endstage renal disease (ESRD). ADPKD is a heterogenic disease which is resulted from the mutations of PKD1 or PKD2 genes which encode polycystin-1 (PC1) and -2 (PC2), thereby multiple cell signaling pathways are involved. METHOD: Although causative genes and aberrant signaling pathways have been investigated for decades, lack of effective and less side-effect treatment for the disease still perplex vast clinicians. Therefore, development of new therapeutic approaches for ADPKD is currently very much desired. CONCLUSION: This review will center on pathogenesis of ADPKD, and thereafter gene transfer will be discussed as potential treatment for the disease. New therapeutic interventions will bring further hope to improve prognosis of this incurable disease.


Subject(s)
Genetic Therapy/trends , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/therapy , Humans , Kidney/pathology , Mutation/genetics , Polycystic Kidney, Autosomal Dominant/pathology , Signal Transduction/genetics , TRPP Cation Channels/genetics , TRPP Cation Channels/therapeutic use
7.
J Cell Mol Med ; 21(8): 1619-1635, 2017 08.
Article in English | MEDLINE | ID: mdl-28244683

ABSTRACT

Although translational research into autosomal dominant polycystic kidney disease (ADPKD) and its pathogenesis has made considerable progress, there is presently lack of standardized animal model for preclinical trials. In this study, we developed an orthologous mouse model of human ADPKD by cross-mating Pkd2 conditional-knockout mice (Pkd2f3 ) to Cre transgenic mice in which Cre is driven by a spectrum of kidney-related promoters. By systematically characterizing the mouse model, we found that Pkd2f3/f3 mice with a Cre transgene driven by the mouse villin-1 promoter (Vil-Cre;Pkd2f3/f3 ) develop overt cysts in the kidney, liver and pancreas and die of end-stage renal disease (ESRD) at 4-6 months of age. To determine whether these Vil-Cre;Pkd2f3/f3 mice were suitable for preclinical trials, we treated the mice with the high-dose mammalian target of rapamycin (mTOR) inhibitor rapamycin. High-dose rapamycin significantly increased the lifespan, lowered the cystic index and kidney/body weight ratio and improved renal function in Vil-Cre;Pkd2f3/f3 mice in a time- and dose-dependent manner. In addition, we further found that rapamycin arrested aberrant epithelial-cell proliferation in the ADPKD kidney by down-regulating the cell-cycle-associated cyclin-dependent kinase 1 (CDK1) and cyclins, namely cyclin A, cyclin B, cyclin D1 and cyclin E, demonstrating a direct link between mTOR signalling changes and the polycystin-2 dysfunction in cystogenesis. Our newly developed ADPKD model provides a practical platform for translating in vivo preclinical results into ADPKD therapies. The newly defined molecular mechanism by which rapamycin suppresses proliferation via inhibiting abnormally elevated CDK1 and cyclins offers clues to new molecular targets for ADPKD treatment.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , CDC2 Protein Kinase/antagonists & inhibitors , Cell Cycle/drug effects , Cyclins/antagonists & inhibitors , Polycystic Kidney, Autosomal Dominant/drug therapy , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cyclins/genetics , Cyclins/metabolism , Dose-Response Relationship, Drug , Female , Founder Effect , Gene Expression Regulation , Humans , Integrases/genetics , Integrases/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Promoter Regions, Genetic , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , TRPP Cation Channels/antagonists & inhibitors , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism
8.
Oncotarget ; 7(41): 67463-67475, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27634899

ABSTRACT

Bladder neck preservation (BNP) during radical prostatectomy (RP) may improve postoperative urinary continence, although its overall effectiveness remains controversial. We systematically searched PubMed, Ovid Medline, Embase, CBM and the Cochrane Library to identify studies published before February 2016 that assessed associations between BNP and post-RP urinary continence. Thirteen trials (1130 cases and 1154 controls) assessing BNP versus noBNP (or with bladder neck reconstruction, BNR) were considered suitable for meta-analysis, including two randomized controlled trials (RCT), six prospective and five retrospective studies. Meta-analysis demonstrated that BNP improved early urinary continence rates (6 mo, OR = 1.66; 95% CI, 1.21-2.27; P = 0.001) and long-term urinary continence outcomes (>12 mo, OR = 3.99; 95% CI, 1.94-8.21; P = 0.0002). Patients with BNP also had lower bladder neck stricture frequencies (OR = 0.49; 95% CI, 0.29-0.81; P = 0.006). Anastomotic leak rates, positive surgical margins and biochemical failure rates were comparable between the two groups (P>0.05). There were no differences in baseline characteristics except for a smaller average prostate volume (WMD = -2.24 ml; 95% CI, -4.27 to -0.22; P = 0.03) in BNP patients. Our analyses indicated that BNP during RP improved early recovery and overall long-term (1 year) urinary continence and decreased bladder neck stricture rates without compromising oncologic control.


Subject(s)
Postoperative Complications/prevention & control , Prostatectomy/methods , Prostatic Neoplasms/surgery , Urinary Bladder/surgery , Urinary Incontinence/prevention & control , Humans , Male , Urinary Incontinence/etiology
9.
Am J Pathol ; 185(10): 2843-60, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26435415

ABSTRACT

Although much is known about the molecular genetic mechanisms of autosomal-dominant polycystic kidney disease (ADPKD), few effective treatment is currently available. Here, we explore the in vivo effects of causal gene replacement in orthologous gene models of ADPKD in mice. Wild-type mice with human PKD2 transgene (PKD2(tg)) overexpressed polycystin (PC)-2 in several tissues, including the kidney and liver, and showed no significant cyst formation in either organ. We cross-mated PKD2(tg) with a Pkd2-null mouse model, which is embryonically lethal and forms renal and pancreatic cysts. Pkd2(-/-) mice with human PKD2 transgene (Pkd2(-/-);PKD2(tg)) were born in expected Mendelian ratios, indicating that the embryonic lethality of the Pkd2(-/-) mice was rescued. Pkd2(-/-);PKD2(tg) mice survived up to 12 months and exhibited moderate to severe cystic phenotypes of the kidney, liver, and pancreas. Moreover, Pkd2(-/-) mice with homozygous PKD2(tg)-transgene alleles (Pkd2(-/-);PKD2(tg/tg)) showed significant further amelioration of the cystic severity compared to that in Pkd2(-/-) mice with a hemizygous PKD2(tg) allele (Pkd2(-/-);PKD2(tg)), suggesting that the ADPKD phenotype was improved by increased transgene dosage. On further analysis, cystic improvement mainly resulted from reduced proliferation, rather apoptosis, of cyst-prone epithelial cells in the mouse model. The finding that the functional restoration of human PC2 significantly rescued ADPKD phenotypes in a dose-dependent manner suggests that increasing PC2 activity may be beneficial in some forms of ADPKD.


Subject(s)
Kidney/pathology , Mutation/genetics , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics , Animals , Cell Proliferation/genetics , Cysts/genetics , Disease Models, Animal , Humans , Kidney/metabolism , Mice, Knockout , Mice, Transgenic , Phenotype
10.
Chin J Cancer Res ; 27(1): 66-73, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25717228

ABSTRACT

OBJECTIVE: The study evaluated the effectiveness of autologous hematopoietic stem cell transplantation (AHSCT) in the treatment of lymphoblastic lymphoma (LL). METHODS: We retrospectively analyzed the data from 41 patients with chemotherapy-sensitive LL who underwent hematopoietic stem cell transplantation (HSCT) from December 1989 to December 2009 in a single institution. RESULTS: HSCT was conducted as first-line consolidation therapy and salvage therapy in 36 and 5 patients, respectively. The median follow-up was 97.1 months (range, 24.6-173.1 months). The 5-year overall survival (OS) and event-free survival (EFS) rate were 64% and 47% for the initially treated patients, respectively, and were both 20% for the relapsed ones. Bone marrow (BM) involvement and chemotherapy cycles prior to transplantation were identified as significant prognostic factors for EFS in multivariate analysis. CONCLUSIONS: These results confirm that AHSCT is a reasonable option for chemotherapy-sensitive LL patients in first complete remission (CR1).

11.
PLoS One ; 9(3): e88816, 2014.
Article in English | MEDLINE | ID: mdl-24594709

ABSTRACT

Bicc1 is a mouse homologue of Drosophila Bicaudal-C (dBic-C), which encodes an RNA-binding protein. Orthologs of dBic-C have been identified in many species, from C. elegans to humans. Bicc1-mutant mice exhibit a cystic phenotype in the kidney that is very similar to human polycystic kidney disease. Even though many studies have explored the gene characteristics and its functions in multiple species, the developmental profile of the Bicc1 gene product (Bicc1) in mammal has not yet been completely characterized. To this end, we generated a polyclonal antibody against Bicc1 and examined its spatial and temporal expression patterns during mouse embryogenesis and organogenesis. Our results demonstrated that Bicc1 starts to be expressed in the neural tube as early as embryonic day (E) 8.5 and is widely expressed in epithelial derivatives including the gut and hepatic cells at E10.5, and the pulmonary bronchi at E11.5. In mouse kidney development, Bicc1 appears in the early ureteric bud and mesonephric tubules at E11.5 and is also expressed in the metanephros at the same stage. During postnatal kidney development, Bicc1 expression gradually expands from the cortical to the medullary and papillary regions, and it is highly expressed in the proximal tubules. In addition, we discovered that loss of the Pkd1 gene product, polycystin-1 (PC1), whose mutation causes human autosomal dominant polycystic kidney disease (ADPKD), downregulates Bicc1 expression in vitro and in vivo. Our findings demonstrate that Bicc1 is developmentally regulated and reveal a new molecular link between Bicc1 and Pkd1.


Subject(s)
RNA-Binding Proteins/genetics , TRPP Cation Channels/physiology , Animals , Base Sequence , Blotting, Western , Cells, Cultured , DNA Primers , Down-Regulation , Immune Sera , Kidney/cytology , Kidney/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , Polymerase Chain Reaction , RNA-Binding Proteins/immunology , TRPP Cation Channels/genetics
12.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 29(7): 723-8, 2013 Jul.
Article in Chinese | MEDLINE | ID: mdl-23837983

ABSTRACT

OBJECTIVE: To produce a rabbit polyclonal antibody, mPkd1-Np, against the extracellular portions of polycystin-1 (PC1) in order to explore the functional roles of the PC1 NH2;-terminus. METHODS: Based on hydrophobic/hydrophilic analyses, we chose a cDNA fragment that encodes amino acids 474E-640L on PC1 and amplified it via RT-PCR. The PCR product was then cloned into a prokaryotic expression vector pGEX-GST. After IPTG induction, the antigen mPkd1-N was produced and further purified. A rabbit was immunized with this antigen and its antiserum was collected. The mPkd1-Np antibody was validated to be specific for PC1 protein through Western blotting, immunohistochemistry, and immunofluorescence methods. RESULTS: The prokaryotic expression vector pGEX-mPkd1-N was successfully constructed and mPkd1-N antigen was induced to express in E.coli Rossetta cells. Using this antigen, the polyclonal antibody mPkd1-Np was produced and its specificity for PC1 was proved through biochemistry and cellular assays. CONCLUSION: We successfully produced an anti-PC1 NH2;-terminal polyclonal antibody named mPkd1-Np. The polyclonal antibody provides a platform for further research into PC1 NH2;-terminal function, specifically renal tubulogenesis and its maintenance.


Subject(s)
Antibodies/immunology , Antibodies/isolation & purification , Protein Interaction Domains and Motifs/immunology , TRPP Cation Channels/immunology , Animals , Antibody Specificity/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Gene Order , Genetic Vectors/genetics , Kidney/metabolism , Mice , Open Reading Frames , Protein Interaction Domains and Motifs/genetics , Rabbits , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , TRPP Cation Channels/chemistry , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism
13.
J Clin Invest ; 121(7): 2845-54, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21701066

ABSTRACT

In addition to its role as an essential neurotransmitter, dopamine serves important physiologic functions in organs such as the kidney. Although the kidney synthesizes dopamine through the actions of aromatic amino acid decarboxylase (AADC) in the proximal tubule, previous studies have not discriminated between the roles of extrarenal and intrarenal dopamine in the overall regulation of renal function. To address this issue, we generated mice with selective deletion of AADC in the kidney proximal tubules (referred to herein as ptAadc-/- mice), which led to selective decreases in kidney and urinary dopamine. The ptAadc-/- mice exhibited increased expression of nephron sodium transporters, decreased natriuresis and diuresis in response to l-dihydroxyphenylalanine, and decreased medullary COX-2 expression and urinary prostaglandin E2 excretion and developed salt-sensitive hypertension. They had increased renin expression and altered renal Ang II receptor (AT) expression, with increased AT1b and decreased AT2 and Mas expression, associated with increased renal injury in response to Ang II. They also exhibited a substantially shorter life span compared with that of wild-type mice. These results demonstrate the importance of the intrarenal dopaminergic system in salt and water homeostasis and blood pressure control. Decreasing intrarenal dopamine subjects the kidney to unbuffered responses to Ang II and results in the development of hypertension and a dramatic decrease in longevity.


Subject(s)
Dopamine/deficiency , Hypertension/physiopathology , Kidney/metabolism , Longevity/physiology , Aldosterone/metabolism , Animals , Aquaporin 2/genetics , Aquaporin 2/metabolism , Aromatic-L-Amino-Acid Decarboxylases/genetics , Aromatic-L-Amino-Acid Decarboxylases/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dopamine/urine , Kidney/anatomy & histology , Kidney/physiopathology , Kidney Tubules, Proximal/enzymology , Mice , Mice, Knockout , Receptors, Drug/genetics , Receptors, Drug/metabolism , Renin/metabolism , Sodium-Bicarbonate Symporters/genetics , Sodium-Bicarbonate Symporters/metabolism , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 1 , Solute Carrier Family 12, Member 3 , Symporters/genetics , Symporters/metabolism
14.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 27(1): 78-81, 2011 Jan.
Article in Chinese | MEDLINE | ID: mdl-21208571

ABSTRACT

AIM: PKHDL1 (the gene for Polycystic Kidney and Hepatic Disease Like-1) had been recently identified, but characteristics of the gene product, Fibrocystin-L (FPC-L), still remain unknown. We therefore produced a rabbit polyclonal antibody hFL-Np to explore the cellular characteristics of this novel protein. METHODS: Based on the hydrophobic/hydrophilic analyses, chose a cDNA fragment which encodes 633L-768K amino acids of the FPC-L and amplified it by RT-PCR. The PCR product was then cloned into a prokaryotic expression vector pGEX-GST. With IPTG induction, the antigen hFL-N was produced and further purified. A rabbit was immunized with the antigen and its antiserum was collected. Applied Western blot with the polyclonal antiserum hFL-Np and validated the antibody specific for FPC-L protein. In addition, also used immunofluorescence staining with hFL-Np to detect the subcellular distribution in cultured HEK293 cells. RESULTS: The prokaryotic expression vector pGEX-hFL-N was successfully constructed and a hFL-N antigen was produced in E.coli Rossetta cells. Using the antigen, a polyclonal antibody hFL-Np was produced and the specificity for FPC-L was also proved by biochemistry and cellular assays. Using the antibody, the cellular staining reveals that FPC-L was a cytosolic protein. CONCLUSION: We produced an anti-FPC-L polyclonal antibody hFL-Np. By biochemistry and cellular characterization, proved that the polyclonal antibody hFL-Np is specific for FPC-L and demonstrated FPC-L is a cytosolic protein. The finding provides a platform for further dissecting FPC-L functions in mammalian development.


Subject(s)
Antibodies/immunology , Receptors, Cell Surface/immunology , Animals , Antibody Formation , Antibody Specificity , Blotting, Western/methods , Cloning, Molecular/methods , Fluorescent Antibody Technique/methods , Genetic Vectors , HEK293 Cells , Humans , Male , Rabbits , Receptors, Cell Surface/genetics , Receptors, Cell Surface/isolation & purification , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
15.
Exp Cell Res ; 317(2): 173-87, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20875407

ABSTRACT

Mutations in the PKHD1 gene result in autosomal recessive polycystic kidney disease (ARPKD) in humans. To determine the molecular mechanism of the cystogenesis in ARPKD, we recently generated a mouse model for ARPKD that carries a targeted mutation in the mouse orthologue of human PKHD1. The homozygous mutant mice display hepatorenal cysts whose phenotypes are similar to those of human ARPKD patients. By littermates of this mouse, we developed two immortalized renal collecting duct cell lines with Pkhd1 and two without. Under nonpermissive culture conditions, the Pkhd1(-/-) renal cells displayed aberrant cell-cell contacts and tubulomorphogenesis. The Pkhd1(-/-) cells also showed significantly reduced cell proliferation and elevated apoptosis. To validate this finding in vivo, we examined proliferation and apoptosis in the kidneys of Pkhd1(-/-) mice and their wildtype littermates. Using proliferation (PCNA and Histone-3) and apoptosis (TUNEL and caspase-3) markers, similar results were obtained in the Pkhd1(-/-) kidney tissues as in the cells. To identify the molecular basis of these findings, we analyzed the effect of Pkhd1 loss on multiple putative signaling regulators. We demonstrated that the loss of Pkhd1 disrupts multiple major phosphorylations of focal adhesion kinase (FAK), and these disruptions either inhibit the Ras/C-Raf pathways to suppress MEK/ERK activity and ultimately reduce cell proliferation, or suppress PDK1/AKT to upregulate Bax/caspase-9/caspase-3 and promote apoptosis. Our findings indicate that apoptosis may be a major player in the cyst formation in ARPKD, which may lead to new therapeutic strategies for human ARPKD.


Subject(s)
Apoptosis/genetics , Epithelial Cells/metabolism , Kidney/metabolism , Polycystic Kidney, Autosomal Recessive/genetics , Receptors, Cell Surface/genetics , Animals , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Transformed , Cell Proliferation , Crosses, Genetic , Cysts/genetics , Disease Models, Animal , Genes, cdc , Genotype , Humans , In Vitro Techniques , Kidney Tubules, Collecting/metabolism , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Mutation , Phenotype , Polycystic Kidney, Autosomal Recessive/metabolism , Polycystic Kidney, Autosomal Recessive/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics
16.
Eur J Cell Biol ; 89(6): 428-36, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20219263

ABSTRACT

The Bicaudal-C (Bic-C) gene was originally discovered in Drosophila melanogaster. The gene product Bic-C is thought to serve as an RNA-binding molecule targeting diverse proteins at the post-transcriptional level. Recent research has shown this gene to be conserved in many species, from Caenorhabditis elegans to humans. Disruption of this protein can disturb the normal migration direction of the anterior follicle cell of Drosophila oocytes, while mutation of a mouse Bicc1 (a mouse homologue of Bic-C) results in phenotypes mimicking human hereditary polycystic kidney disease (PKD). However, the cellular function of Bicc1 gene products in mammalian systems remains largely unknown. In this study, we established stable IMCD (mouse inner medullary collecting duct) cell lines, in which Bicc1 was silenced by short hairpin RNA inhibition (shRNA). We show that inhibition of Bicc1 disrupted normal tubulomorphogenesis and induced cystogenesis of IMCD cells grown in three dimensional cultures. To determine what factors contributed to the defect, we systematically examined biological changes of Bicc1-silenced IMCD cells. We found that the cells had significant defects in E-cadherin-based cell-cell adhesion, along with abnormalities in actin cytoskeleton organization, cell-extracellular matrix interactions, cell proliferation, and apoptosis. These findings suggest that lack of Bicc1 leads to disruption of normal cell-cell junctions, which in turn impedes establishment of epithelial polarity. These cellular defects may initiate abnormal tubulomorphogenesis and cystogenesis of IMCD cells grown in vitro. The observation of aberrant cellular behaviors in Bicc1-silenced IMCD cells reveal functions for Bicc1 in renal epithelial cells and provides insight into a potential pathogenic mechanism of polycystic kidney disease.


Subject(s)
Cadherins/metabolism , Carrier Proteins/metabolism , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , Animals , Apoptosis/genetics , Blotting, Western , Carrier Proteins/genetics , Cell Adhesion/genetics , Cell Communication/genetics , Cell Line , Cell Movement/genetics , Cell Polarity/genetics , Cell Proliferation , Fluorescent Antibody Technique , Gene Silencing , Mice , Microscopy, Confocal , RNA-Binding Proteins , Reverse Transcriptase Polymerase Chain Reaction , Transfection
17.
J Am Soc Nephrol ; 20(12): 2556-69, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19939939

ABSTRACT

Loss of polycystin-2 (PC2) in mice (Pkd2(-/-)) results in total body edema, focal hemorrhage, structural cardiac defects, abnormal left-right axis, hepatorenal and pancreatic cysts, and embryonic lethality. The molecular mechanisms by which loss of PC2 leads to these phenotypes remain unknown. We generated a model to allow targeted Pkd2 inactivation using the Cre-loxP system. Global inactivation of Pkd2 produced a phenotype identical to Pkd2(-/-) mice with undetectable PC2 protein and perinatal lethality. Using various Cre mouse lines, we found that kidney, pancreas, or time-specific deletion of Pkd2 led to cyst formation. In addition, we developed an immortalized renal collecting duct cell line with inactive Pkd2; these cells had aberrant cell-cell contact, ciliogenesis, and tubulomorphogenesis. They also significantly upregulated beta-catenin, axin2, and cMyc. Our results suggest that loss of PC2 disrupts normal behavior of renal epithelial cells through dysregulation of beta-catenin-dependent signaling, revealing a potential role for this signaling pathway in PC2-associated ADPKD.


Subject(s)
Mutation , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics , beta Catenin/metabolism , Animals , Apoptosis , Cell Line , Cell Proliferation , Cysts/genetics , Cysts/pathology , Disease Models, Animal , Female , Kidney Tubules, Collecting/abnormalities , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Collecting/pathology , Liver Diseases/genetics , Liver Diseases/pathology , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Diseases/genetics , Pancreatic Diseases/pathology , Phenotype , Polycystic Kidney, Autosomal Dominant/etiology , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Pregnancy , Signal Transduction , TRPP Cation Channels/deficiency , TRPP Cation Channels/metabolism , Up-Regulation
18.
J Biol Chem ; 283(46): 31559-66, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-18782757

ABSTRACT

Autosomal dominant (ADPKD) and autosomal recessive (ARPKD) polycystic kidney disease are caused by mutations in Pkd1/Pkd2 and Pkhd1, which encode polycystins (PCs) and fibrocystin/polyductin (FPC). Our recent study reported that a deficiency in FPC increases the severity of cystic disease in Pkd2 mutants and down-regulates PC2 in vivo, but the precise molecular mechanism of these effects is unknown (Kim, I., Fu, Y., Hui, K., Moeckel, G., Mai, W., Li, C., Liang, D., Zhao, P., Ma, J., Chen, X.-Z., George, A. L., Jr., Coffey, R. J., Feng, Z. P., and Wu, G. (2008) J. Am. Soc. Nephrol. 19, 455-468). In this study, through the use of deletion and mutagenesis strategies, we identified a PC2-binding domain in the intracellular C terminus of FPC and an FPC-binding domain in the intracellular N terminus of PC2. These binding domains provide a molecular basis for the physical interaction between PC2 and FPC. In addition, we also found that physical interaction between the binding domains of PC2 and FPC is able to prevent down-regulation of PC2 induced by loss of FPC. In vivo, we generated a mouse model of ADPKD with hypomorphic Pkd2 alleles (Pkd2nf3/nf3) and show that PC2 down-regulation is accompanied by a phenotype similar to that of Pkhd1(-/-) mice. These findings demonstrate a common mechanism underlying cystogenesis in ADPKD and ARPKD and provide insight into the molecular relationship between PC2 and FPC.


Subject(s)
Receptors, Cell Surface/metabolism , TRPP Cation Channels/metabolism , Alleles , Amino Acid Sequence , Animals , Down-Regulation , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Phenotype , Protein Binding , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Sequence Alignment , Sequence Homology, Amino Acid , TRPP Cation Channels/chemistry , TRPP Cation Channels/genetics
19.
J Am Soc Nephrol ; 19(3): 455-68, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18235088

ABSTRACT

Autosomal recessive polycystic kidney disease is caused by mutations in PKHD1, which encodes the membrane-associated receptor-like protein fibrocystin/polyductin (FPC). FPC associates with the primary cilia of epithelial cells and co-localizes with the Pkd2 gene product polycystin-2 (PC2), suggesting that these two proteins may function in a common molecular pathway. For investigation of this, a mouse model with a gene-targeted mutation in Pkhd1 that recapitulates phenotypic characteristics of human autosomal recessive polycystic kidney disease was produced. The absence of FPC is associated with aberrant ciliogenesis in the kidneys of Pkhd1-deficient mice. It was found that the COOH-terminus of FPC and the NH2-terminus of PC2 interact and that lack of FPC reduced PC2 expression but not vice versa, suggesting that PC2 may function immediately downstream of FPC in vivo. PC2-channel activities were dysregulated in cultured renal epithelial cells derived from Pkhd1 mutant mice, further supporting that both cystoproteins function in a common pathway. In addition, mice with mutations in both Pkhd1 and Pkd2 had a more severe renal cystic phenotype than mice with single mutations, suggesting that FPC acts as a genetic modifier for disease severity in autosomal dominant polycystic kidney disease that results from Pkd2 mutations. It is concluded that a functional and molecular interaction exists between FPC and PC2 in vivo.


Subject(s)
Kidney Tubules/pathology , Polycystic Kidney, Autosomal Recessive/metabolism , Receptors, Cell Surface/metabolism , TRPP Cation Channels/metabolism , Animals , Cells, Cultured , Cilia/pathology , Disease Models, Animal , Disease Progression , Down-Regulation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/physiology , Humans , Ion Channels/metabolism , Kidney Tubules/metabolism , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Phenotype , Polycystic Kidney, Autosomal Recessive/genetics , Polycystic Kidney, Autosomal Recessive/pathology , Receptors, Cell Surface/genetics , Urothelium/metabolism , Urothelium/pathology
20.
Hum Mol Genet ; 17(8): 1109-19, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18178578

ABSTRACT

Endoplasmic reticulum(ER)-associated degradation (ERAD) is an essential process for cell homeostasis and remains not well understood. During ERAD, misfolded proteins are recognized, ubiquitinated on ER and subsequently retro-translocated/dislocated from ER to the 26S proteasome in the cytosol for proteolytic elimination. Polycystin-2 (PC2), a member of the transient receptor potential superfamily of cation channels, is a Ca channel mainly located on ER and primary cilium membranes of cells. Mutations in PC2 are associated with autosomal dominant polycystic kidney disease (ADPKD). The cellular and molecular mechanisms underlying the PC2-associated pathogenesis remain unclear. Here we show that PC2 degradation is regulated by the ERAD pathway through the ubiquitin-proteasome system. PC2 interacted with ATPase p97, a well-known ERAD component extracting substrates from ER, and immobilized it in perinuclear regions. PC2 also interacted with Herp, an ubiquitin-like protein implicated in regulation of ERAD. We found that Herp is required for and promotes PC2 degradation. ER stress accelerates the retro-translocation of PC2 for cytosolic degradation, at least in part through increasing the Herp expression. Thus, PC2 is a novel ERAD substrate. Herp also promoted, to varied degrees, the degradation of PC2 truncation mutants, including two pathogenic mutants R872X and E837X, as long as they interact with Herp. In contrast, Herp did not interact with, and has no effect on the degradation of, PC2 mutant missing both the N- and C-termini. The ERAD machinery may thus be important for ADPKD pathogenesis because the regulation of PC2 expression by the ERAD pathway is altered by mutations in PC2.


Subject(s)
Endoplasmic Reticulum/metabolism , TRPP Cation Channels/metabolism , Animals , Cell Line , Dogs , Humans , Membrane Proteins/metabolism , Metabolic Networks and Pathways , Mutation , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...