85 results on '"Cebotaru L"'
Search Results
2. 604: Transduction of Rhesus macaque lung after repeat dosing by AAV1 is enhanced by short-term prednisone treatment
- Author
-
Yanda, M., primary, Tomar, V., additional, Cebotaru, C., additional, Guggino, W., additional, and Cebotaru, L., additional
- Published
- 2021
- Full Text
- View/download PDF
3. WNK4 kinase regulates surface expression of the human sodium chloride cotransporter in mammalian cells
- Author
-
Cai, H, Cebotaru, V, Wang, Y-H, Zhang, X-M, Cebotaru, L, Guggino, S E, and Guggino, W B
- Published
- 2006
4. P025 The PDZ-containing domain protein, CAL links CFTR to the regulation of inflammation
- Author
-
Cebotaru, L., primary and Guggino, W., additional
- Published
- 2019
- Full Text
- View/download PDF
5. EPS3.01 Transduction of rhesus macaque lung following repeat dosing by adeno-associated virus serotype 1
- Author
-
Yanda, M.K., primary, Wang, H., additional, Guggino, W.B., additional, and Cebotaru, L., additional
- Published
- 2018
- Full Text
- View/download PDF
6. WS15.2 R334W CFTR, a severely compromised chloride conductance mutant, retains its bicarbonate conductance and responds to the corrector combination, C4 + C18
- Author
-
Boinot, C., primary, Wang, H., additional, Sabirzhanova, I., additional, Cebotaru, C., additional, Guggino, W., additional, and Cebotaru, L., additional
- Published
- 2018
- Full Text
- View/download PDF
7. 32 CFTR NBD2 mutant N1303K associates with and influences the functioning of the autophagosome
- Author
-
Sabirzhanova, S., primary, Boinot, C., additional, and Cebotaru, L., additional
- Published
- 2017
- Full Text
- View/download PDF
8. EPS1.2 Transduction of rhesus macaque lung by AAV1
- Author
-
Guggino, W.B., primary, Engelhardt, J., additional, Yan, Z., additional, and Cebotaru, L., additional
- Published
- 2017
- Full Text
- View/download PDF
9. 10 Gene therapy to rescue of premature stop mutations R1162X and W1282X
- Author
-
Cebotaru, L., primary, Boinot, C., additional, Sabirzhanova, I., additional, and Guggino, W.B., additional
- Published
- 2016
- Full Text
- View/download PDF
10. 33 Rescue of premature stop mutations R1162X and W1282X by a combination of small molecule correctors
- Author
-
Cebotaru, L., primary, Boinot, C., additional, Sabirzhanova, I., additional, and Guggino, W.B., additional
- Published
- 2016
- Full Text
- View/download PDF
11. WS15.5 Transcomplementation by a truncation mutant of CFTR, Δ27-264 CFTR, rescues both trafficking and chloride channel function of ΔF508 CFTR
- Author
-
Cebotaru, L., primary, Woodward, O., additional, and Guggino, W., additional
- Published
- 2012
- Full Text
- View/download PDF
12. CFTR missing the last four transmembrane segments and nucleotide binding domain 2 resides in the ER but escapes the cell's quality control mechanism
- Author
-
Cebotaru, L., primary and Guggino, W.B., additional
- Published
- 2010
- Full Text
- View/download PDF
13. Rescue of A455E CFTR by temperature, small molecule correctors and transcomplementation
- Author
-
Cebotaru, L., primary and Guggino, W., additional
- Published
- 2009
- Full Text
- View/download PDF
14. CFTR missing the first four transmembrane segments enhances wild type and ΔF508 CFTR processing
- Author
-
Cebotaru, L., primary, Vij, N., additional, Ciobanu, I., additional, Wright, J., additional, and Guggino, W., additional
- Published
- 2008
- Full Text
- View/download PDF
15. 56 AAV Δ264CFTR enhances wt CFTR expression and maturation of ΔF508CFTR
- Author
-
Cebotaru, L., primary, Flotté, T.R., additional, and Guggino, W., additional
- Published
- 2006
- Full Text
- View/download PDF
16. Transduction of Ferret Surface and Basal Cells of Airways, Lung, Liver, and Pancreas via Intratracheal or Intravenous Delivery of Adeno-Associated Virus 1 or 6.
- Author
-
Yanda MK, Zeidan A, Ciobanu C, Izzi J, Guggino WB, and Cebotaru L
- Subjects
- Animals, Dependovirus genetics, Lung, Liver, Pancreas, RNA, Messenger, Genetic Vectors genetics, Transduction, Genetic, Ferrets genetics, Cystic Fibrosis
- Abstract
Cystic fibrosis (CF) is potentially treatable by gene therapy. Since the identification of the CF gene, preclinical and clinical trials have concentrated on achieving effective gene therapy targeting the lung. However, the lung has proven to be a formidable barrier to successful gene therapy especially for CF, and many clinical trials failed to achieve efficacy. Recent advances in vector design and adeno-associated virus (AAV) serotypes have increased the chances of success. Given that CF is a multi-organ disease, the goal of this study was to test whether a gene therapy approach involving AAV1 or AAV6 vector delivery via the systemic circulation would at the same time overcome the barrier of lung delivery and transduce organs commonly affected by CF. To accomplish this, we sprayed AAV1 containing green fluorescent protein (GFP) into the trachea or injected it intravenously (IV). We also tested AAV6 injected IV. No adverse events were noted. Ferrets were necropsied 30 days after vector delivery. AAV1 or AAV6 vector genomes, messenger RNA (mRNA) expression, and GFP were detected in all the tracheal and lung samples from the treated animals, whether AAV1 was sprayed into the trachea or injected IV or AAV6 was injected IV. Importantly, both surface epithelial and basal cells of the trachea and lung airways were successfully transduced, regardless of which route of delivery or vector serotype used for transduction. We detected also AAV1 and AAV6 vector genomes, mRNA expression, and GFP in the livers and pancreases, particularly in the acinar cells of the pancreatic duct. These data suggest that gene transfer is attainable in the airways, liver, and pancreas using either serotype, AAV1 or AAV6. Given that these same organs are affected in CF, systemic delivery of AAV may be the preferred route of delivery for a gene therapy for CF.
- Published
- 2023
- Full Text
- View/download PDF
17. CFTR and PC2, partners in the primary cilia in autosomal dominant polycystic kidney disease.
- Author
-
Yanda MK, Ciobanu C, Guggino WB, and Cebotaru L
- Subjects
- Animals, Mice, Cilia metabolism, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Kidney metabolism, Septins genetics, Septins metabolism, Polycystic Kidney, Autosomal Dominant genetics, Polycystic Kidney, Autosomal Dominant metabolism
- Abstract
Defects in the primary cilium are associated with autosomal dominant polycystic kidney disease (ADPKD). We used a combination of animal models, Western blotting, and confocal microscopy and discovered that CFTR and polycystin 2 (PC2) are both colocalized to the cilium in normal kidneys, with the levels of both being decreased in cystic epithelia. Cilia were longer in CFTR-null mice and in cystic cells in our ADPKD animal models. We examined septin 2, known to play a role in cilia length, to act as a diffusion barrier and to serve as an enhancer of proliferation. We found that septin 2 protein levels were upregulated and colocalized strongly with CFTR in cystic cells. Application of VX-809, the CFTR corrector, restored CFTR and PC2 toward normal in the cilia, decreased the protein levels of septin 2, and drastically reduced septin 2 colocalization with CFTR. Our data suggest that CFTR is present in the cilia and plays a role there, perhaps through its conductance of Cl
- . We also postulate that septin 2 is important for localizing CFTR to the apical membrane in cystic epithelia. NEW & NOTEWORTHY CFTR is present in the primary cilia together with polycystin 2 (PC2). Ablation of CFTR makes cilia longer suggesting that CFTR plays a role there, perhaps through its conductance of Cl.- Published
- 2023
- Full Text
- View/download PDF
18. Ameliorating liver disease in an autosomal recessive polycystic kidney disease mouse model.
- Author
-
Yanda MK, Zeidan A, and Cebotaru L
- Subjects
- Humans, Mice, Animals, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Receptors, Cell Surface metabolism, Liver Cirrhosis complications, Heat-Shock Proteins metabolism, Polycystic Kidney, Autosomal Recessive drug therapy, Polycystic Kidney, Autosomal Recessive genetics, Polycystic Kidney, Autosomal Recessive metabolism
- Abstract
Systemic and portal hypertension, liver fibrosis, and hepatomegaly are manifestations associated with autosomal recessive polycystic kidney disease (ARPKD), which is caused by malfunctions of fibrocystin/polyductin (FPC). The goal is to understand how liver pathology occurs and to devise therapeutic strategies to treat it. We injected 5-day-old Pkhd1
del3-4/del3-4 mice for 1 mo with the cystic fibrosis transmembrane conductance regulator (CFTR) modulator VX-809 designed to rescue processing and trafficking of CFTR folding mutants. We used immunostaining and immunofluorescence techniques to evaluate liver pathology. We assessed protein expression via Western blotting. We detected abnormal biliary ducts consistent with ductal plate abnormalities, as well as a greatly increased proliferation of cholangiocytes in the Pkhd1del3-4/del3-4 mice. CFTR was present in the apical membrane of cholangiocytes and increased in the Pkhd1del3-4/del3-4 mice, consistent with a role for apically located CFTR in enlarged bile ducts. Interestingly, we also found CFTR in the primary cilium, in association with polycystin (PC2). Localization of CFTR and PC2 and overall length of the cilia were increased in the Pkhd1del3-4/del3-4 mice. In addition, several of the heat shock proteins; 27, 70, and 90 were upregulated, suggesting that global changes in protein processing and trafficking had occurred. We found that a deficit of FPC leads to bile duct abnormalities, enhanced cholangiocyte proliferation, and misregulation of heat shock proteins, which all returned toward wild type (WT) values following VX-809 treatment. These data suggest that CFTR correctors can be useful as therapeutics for ARPKD. Given that these drugs are already approved for use in humans, they can be fast-tracked for clinical use. NEW & NOTEWORTHY ARPKD is a multiorgan genetic disorder resulting in newborn morbidity and mortality. There is a critical need for new therapies to treat this disease. We show that persistent cholangiocytes proliferation occurs in a mouse model of ARPKD along with mislocalized CFTR and misregulated heat shock proteins. We found that VX-809, a CFTR modulator, inhibits proliferation and limits bile duct malformation. The data provide a therapeutic pathway for strategies to treat ADPKD.- Published
- 2023
- Full Text
- View/download PDF
19. Short-Term Steroid Treatment of Rhesus Macaque Increases Transduction.
- Author
-
Yanda MK, Tomar V, Cebotaru CV, Guggino WB, and Cebotaru L
- Subjects
- Animals, Dependovirus metabolism, Genetic Vectors genetics, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Macaca mulatta genetics, Macaca mulatta metabolism, Methylprednisolone pharmacology, Methylprednisolone therapeutic use, Steroids, Cystic Fibrosis genetics, Cystic Fibrosis Transmembrane Conductance Regulator genetics
- Abstract
Repeat dosing poses a major hurdle for the development of an adeno-associated virus (AAV)-based gene therapy for cystic fibrosis, in part because of the potential for development of an immune reaction to the AAV1 capsid proteins. Here, to dampen the immune response to AAV1, we treated Rhesus monkeys with methylprednisolone before and after the instillation of two doses of AAV1Δ27-264-CFTR into their airways at 0 and 30 days, followed by a single dose of AAV1-GFP on day 60. Animals were euthanized on day 90, except for one monkey that was sacrificed at 1 year. No adverse events occurred, indicating that the two AAV1 vectors are safe. rAAV1-CFTR and AAV1-GFP vector genomes and mRNA transcripts were detectable in all lung sections and in the liver and pancreas at day 90 and after 1 year at levels comparable with animals necropsied at 90 days. The numbers of vector genomes for cystic fibrosis transmembrane regulator (CFTR) and green fluorescent protein (GFP) detected here were higher than those found in the monkeys infected without methylprednisolone treatment that we tested previously.
1 Also, lung surface and keratin 5-positive basal cells showed higher CFTR and GFP staining than did the cells from the uninfected monkey control. Positive immunostaining, also detected in the liver and pancreas, remained stable for at least a year. All animals seroconverted for anticapsid antibodies by 90 days post-treatment. The neutralizing antibody titer declined in the animal necropsied at 1 year. Conclusion: AAV1 safely and effectively transduces monkey airway and basal cells. Both the presence of vector genomes and transduction from AAV1-CFTR and AAV1-GFP virus seen in the monkeys 4 months to 1 year after the first instillation suggest that repeat dosing with AAV1-based vectors is achievable, particularly after methylprednisolone treatment.- Published
- 2022
- Full Text
- View/download PDF
20. Therapeutic Potential for CFTR Correctors in Autosomal Recessive Polycystic Kidney Disease.
- Author
-
Yanda MK, Tomar V, and Cebotaru L
- Subjects
- Animals, Cell Line, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Disease Models, Animal, Endoplasmic Reticulum-Associated Degradation, Gene Expression Regulation, Gene Silencing, Genetic Therapy methods, Heat-Shock Proteins genetics, Heat-Shock Proteins metabolism, Humans, Mice, Mice, Knockout, Mutation, Phenotype, Polycystic Kidney, Autosomal Recessive diagnosis, Polycystic Kidney, Autosomal Recessive therapy, Protein Transport, Receptors, Cell Surface metabolism, Sequence Deletion, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Genetic Predisposition to Disease, Polycystic Kidney, Autosomal Recessive genetics, Polycystic Kidney, Autosomal Recessive metabolism, Receptors, Cell Surface genetics
- Abstract
Background & Aims: Autosomal recessive polycystic kidney disease (ARPKD) is caused by mutations in PKHD1, encoding fibrocystin/polyductin (FPC). Severe disease occurs in perinates. Those who survive the neonatal period face a myriad of comorbidities, including systemic and portal hypertension, liver fibrosis, and hepatosplenomegaly. The goal here was to uncover therapeutic strategies for ARPKD., Methods: We used wild-type and an FPC-mutant cholangiocyte cell line in 3-dimenional cysts and in confluent monolayers to evaluate protein expression using western blotting and protein trafficking using confocal microscopy., Results: We found that the protein level of the cystic fibrosis transmembrane conductance regulator (CFTR) was downregulated. The levels of heat shock proteins (HSPs) were altered in the FPC-mutant cholangiocytes, with HSP27 being downregulated and HSP90 and HSP70 upregulated. FPC-mutant cholangiocytes formed cysts, but normal cells did not. Cyst growth could be reduced by increasing HSP27 protein levels, by HSP90 and HSP70 inhibitor treatments, by silencing HSP90 through messenger RNA inhibition, or by the novel approach of treating the cysts with the CFTR corrector VX-809. In wild-type cholangiocytes, CFTR is present in both apical and basolateral membranes. FPC malfunction resulted in altered colocalization of CFTR with both apical and basolateral membranes. Whereas, treatment with VX-809, increasing HSP27 or inhibiting HSP70 or HSP90 restored CFTR localization toward normal values., Conclusions: FPC malfunction induces the formation of cysts, which are fueled by alterations in HSPs and in CFTR protein levels and miss-localization. We suggest that CFTR correctors, already in clinical use to treat cystic fibrosis, could also be used as a treatment for ARPKD., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
21. Transduction of Surface and Basal Cells in Rhesus Macaque Lung Following Repeat Dosing with AAV1CFTR.
- Author
-
Guggino WB, Yanda MK, Cebotaru CV, and Cebotaru L
- Subjects
- Animals, Cystic Fibrosis genetics, Cystic Fibrosis immunology, Dose-Response Relationship, Drug, Gene Transfer Techniques, Genetic Therapy, Genetic Vectors genetics, Genome, Viral, Humans, Lung immunology, Lung pathology, Macaca mulatta, Cystic Fibrosis therapy, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Dependovirus genetics, Genetic Vectors administration & dosage, Lung metabolism, Transduction, Genetic
- Abstract
To test the effectiveness of repeat dosing, we sprayed two doses (10
13 vg each) of AAV1Δ27-264-CFTR into airways of four rhesus monkeys at 0 and 30 days, followed by a single dose of 1013 vg of AAV1GFP on day 60. Monkeys were sacrificed on day 90. No adverse events occurred, indicating that AAV1 vectors are safe. An elevated anti-AAV1 neutralizing titer was established by the third dose. A positive ELISPOT to the adeno-associated virus (AAV) capsid but not to cystic fibrosis transmembrane conductance regulator (CFTR) occurred after the third dose in three monkeys. AAV1-CFTR and GFP vectors were detectable in all lung sections and in the heart, liver, and spleen. The CFTR protein was higher in treated monkeys than in an untreated monkey. GFP protein was detected in treated lungs. Lung surface and keratin 5-positive basal cells showed higher CFTR staining than in the uninfected monkey and were positive for GFP staining, indicating widespread gene transduction by AAV1CFTR and GFP. AAV1 safely and effectively transduces monkey airway and basal cells. Both the significant numbers of vector genomes and transduction from AAV1CFTR and GFP virus seen in the monkeys 3 months after the first instillation suggest that repeat dosing with AAV1-based vectors is achievable.- Published
- 2020
- Full Text
- View/download PDF
22. Gene Therapy for Cystic Fibrosis Paved the Way for the Use of Adeno-Associated Virus in Gene Therapy.
- Author
-
Guggino WB and Cebotaru L
- Subjects
- Animals, Gene Transfer Techniques, Genetic Vectors, Humans, Promoter Regions, Genetic, Respiratory System virology, Cystic Fibrosis genetics, Cystic Fibrosis therapy, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Dependovirus genetics, Genetic Therapy
- Abstract
Shortly after the cystic fibrosis (CF) gene was identified in 1989, the race began to develop a gene therapy for this condition. Major efforts utilized full-length cystic fibrosis transmembrane conductance regulator packaged into adenovirus, adeno-associated virus (AAV), or liposomes and delivered to the airways. The drive to find a treatment for CF based on gene therapy drove the early stages of gene therapy in general, particularly those involving AAV gene therapy. Since general overviews of CF gene therapy have already been published, this review considers specifically the efforts using AAV and is focused on honoring the contributions of Dr. Barrie Carter.
- Published
- 2020
- Full Text
- View/download PDF
23. A new role for heat shock factor 27 in the pathophysiology of Clostridium difficile toxin B.
- Author
-
Yanda MK, Guggino WB, and Cebotaru L
- Subjects
- Caco-2 Cells, Chlorides metabolism, Clostridioides difficile pathogenicity, Clostridium Infections microbiology, Clostridium Infections physiopathology, Colon microbiology, Colon physiopathology, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Diarrhea microbiology, Diarrhea physiopathology, Electric Impedance, Heat-Shock Proteins genetics, Host-Pathogen Interactions, Humans, Molecular Chaperones genetics, Permeability, Protein Binding, Signal Transduction, Tissue Culture Techniques, Bacterial Proteins metabolism, Bacterial Toxins metabolism, Clostridioides difficile metabolism, Clostridium Infections metabolism, Colon innervation, Diarrhea metabolism, Heat-Shock Proteins metabolism, Molecular Chaperones metabolism
- Abstract
Clostridium difficile (CD) is a common pathogen that causes severe gastrointestinal inflammatory diarrhea in patients undergoing antibiotic therapy. Its virulence derives from two toxins, toxin CD, A and B (TcdA and TcdB) (Borriello et al. Rev Infect Dis 12, Suppl 2: S185-191, 1990). Among the prime candidates for CD colonization are patients with cystic fibrosis (CF), who are routinely treated with antibiotics and frequently hospitalized. Indeed, ~50% of patients with CF are colonized with virulent forms of CD but do not exhibit diarrhea (Bauer et al. Clin Microbiol Infect 20: O446-O449, 2014; Binkovitz et al. Am J Roentgenol 172: 517-521, 199; Zemljic et al. Anaerobe 16: 527-532, 2010). We found that TcdB has global effects on colonic cells, including reducing the steady-state levels of sodium-proton exchange regulatory factors, reducing the levels of heat shock protein (Hsp) 27, and increasing the fraction of total Hsp27 bound to the cystic fibrosis transmembrane conductance regulator (CFTR). Also, since some mutations in CFTR seem to be protective, we asked whether CFTR is a target of TcdB. We show here that TcdB increases the maturation of CFTR and transiently increases its function. These combined effects promote increased surface expression of CFTR, resulting in a transient increase in Cl
- secretion. This increase is followed by a precipitous decline in both CFTR-dependent Cl- secretion and transepithelial resistance (TER), suggesting a breakdown in the epithelial cells' tight junctions. We also found that overexpressing Hsp27 reverses some of the deleterious effects of TcdB, in particular preserving TER and therefore likely the maintenance of barrier function. Thus, our data suggest that Hsp27 plays a role in the diarrhea generated by CD infection and is a potential therapeutic target for treating this diarrhea. NEW & NOTEWORTHY Clostridium difficile (CD) is a common pathogen that causes severe gastrointestinal inflammatory diarrhea in patients undergoing antibiotic therapy. We provide new evidence that heat shock protein (Hsp) 27 is one of the key players in CD pathology and that increasing Hsp27 can prevent the decrease in transepithelial resistance induced by toxin CD B, pointing the way for pharmacologic therapies for patients with chronic CD infection that can increase Hsp27 as a means to mitigate the effects of CD on gastrointestinal pathology.- Published
- 2020
- Full Text
- View/download PDF
24. Pharmacological reversal of renal cysts from secretion to absorption suggests a potential therapeutic strategy for managing autosomal dominant polycystic kidney disease.
- Author
-
Yanda MK, Cha B, Cebotaru CV, and Cebotaru L
- Subjects
- Absorption, Physicochemical, Aminopyridines pharmacology, Aminopyridines therapeutic use, Benzodioxoles pharmacology, Benzodioxoles therapeutic use, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Endoplasmic Reticulum drug effects, Endoplasmic Reticulum metabolism, Golgi Apparatus drug effects, Golgi Apparatus metabolism, Humans, Polycystic Kidney, Autosomal Dominant complications, Polycystic Kidney, Autosomal Dominant pathology, Protein Transport drug effects, Sodium-Hydrogen Exchanger 3 metabolism, Cysts complications, Polycystic Kidney, Autosomal Dominant drug therapy, Polycystic Kidney, Autosomal Dominant metabolism
- Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) induces a secretory phenotype, resulting in multiple fluid-filled cysts. We have previously demonstrated that VX-809, a corrector of the cystic fibrosis transmembrane conductance regulator (CFTR), reduces cyst growth. Here, we show that in normal mice CFTR is located within the cells and also at the apical and basolateral membranes. However, in polycystic kidney disease ( pkd1 )-knockout mice, CFTR was located at the plasma membrane, consistent with its role in cAMP-dependent fluid secretion. In cystic mice, VX-809 treatment increased CFTR levels at the apical membrane and reduced its association with the endoplasmic reticulum. Surprisingly, VX-809 treatment significantly increased CFTR's co-localization with the basolateral membrane in cystic mice. Na
+ /H+ exchanger 3 (NHE3) is present in pkd1 -knockout and normal mice and in proximal tubule-derived, cultured pkd1 -knockout cells. VX-809 increased the expression, activity, and apical plasma membrane localization of NHE3. Co-localization of epithelial sodium channel (ENaC) with the plasma membrane was reduced in cysts in pkd1 -knockout mice, consistent with an inability of the cysts to absorb fluid. Interestingly, in the cystic mice, VX-809 treatment increased ENaC levels at the apical plasma membrane consistent with fluid absorption. Thus, VX-809 treatment of pkd1 -null mouse kidneys significantly affected CFTR, NHE3, and ENaC, altering the cyst phenotype from one poised toward fluid secretion toward one more favorable for absorption. VX-809 also altered the location of CFTR but not of NHE3 or ENaC in normal mice. Given that VX-809 administration is safe, it may have potential utility for treating patients with ADPKD., (© 2019 Yanda et al.)- Published
- 2019
- Full Text
- View/download PDF
25. The CFTR Corrector, VX-809 (Lumacaftor), Rescues ABCA4 Trafficking Mutants: a Potential Treatment for Stargardt Disease.
- Author
-
Liu Q, Sabirzhanova I, Bergbower EAS, Yanda M, Guggino WG, and Cebotaru L
- Subjects
- ATP-Binding Cassette Transporters genetics, Aminopyridines therapeutic use, Anilides pharmacology, Benzodioxoles therapeutic use, Cell Membrane metabolism, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Gene Expression Regulation drug effects, HEK293 Cells, HSP27 Heat-Shock Proteins metabolism, Humans, Hydroxamic Acids pharmacology, Leupeptins pharmacology, Lysosomes metabolism, Macular Degeneration congenital, Macular Degeneration drug therapy, Macular Degeneration metabolism, Macular Degeneration pathology, Mutation, Protein Transport drug effects, Stargardt Disease, ATP-Binding Cassette Transporters metabolism, Aminopyridines pharmacology, Benzodioxoles pharmacology
- Abstract
Background/aims: Mutations in ABCA4 cause Stargardt macular degeneration, which invariably ends in legal blindness. We studied two common mutants, A1038V (in NBD1) and G1961E (in NBD2), with the purpose of exploring how they interact with the cell's quality control mechanism. The study was designed to determine how these mutants can be rescued., Methods: We expressed wt and mutant ABCA4 in HEK293 cells and studied the effect of the mutations on trafficking and processing and the ability of correctors to rescue them. We used a combination of western blotting, confocal microscopy and surface biotinylation coupled with pulldown of plasma membrane proteins., Results: G1961E is sensitive to inhibitors of the aggresome, tubacin and the lysosome, bafilomycin A. Both mutants cause a reduction in heat shock protein, Hsp27. Incubation of HEK293 cells expressing the mutants with VX-809, an FDA approved drug for the treatment of cystic fibrosis, increased the levels of A1038V and G1961E by 2- to 3-fold. Importantly, VX-809 increased the levels of both mutants at the plasma membrane suggesting that trafficking had been restored. Transfecting additional Hsp27 to the cells also increased the steady state levels of both mutants. However, in combination with VX-809 the addition of Hsp27 caused a dramatic increase in the protein expression particularly in the G1961 mutant which increased approximately 5-fold., Conclusion: Our results provide a new mechanism for the rescue of ABCA4 trafficking mutants based on the restoration of Hsp27. Our results provide a pathway for the treatment of Stargardt disease., Competing Interests: The authors declare they have no conflict of interest., (© Copyright by the Author(s). Published by Cell Physiol Biochem Press.)
- Published
- 2019
- Full Text
- View/download PDF
26. Restoration of F508-del Function by Transcomplementation: The Partners Meet in the Endoplasmic Reticulum.
- Author
-
Bergbower EAS, Sabirzhanova I, Boinot C, Guggino WB, and Cebotaru L
- Subjects
- Cell Line, Cystic Fibrosis therapy, Genetic Therapy, Humans, Respiratory Mucosa cytology, Respiratory Mucosa metabolism, Sequence Deletion, Transfection, Cystic Fibrosis genetics, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Dependovirus genetics, Endoplasmic Reticulum genetics
- Abstract
Background/aims: Because of the small size of adeno-associated virus, AAV, the cystic fibrosis conductance regulator, CFTR, cDNA is too large to fit within AAV and must be truncated. We report here on two truncated versions of CFTR, which, when inserted into AAV1 and used to infect airway cells, rescue F508-del CFTR via transcomplementation. The purpose of this study is to shed light on where in the cell transcomplementation occurs and how it results in close association between the endogenous F508-del and truncated CFTR., Methods: We treated CF airway cells (CFBE41o
- ) with AAV2/1 (AAV2 inverted terminal repeats/AAV1 capsid) containing truncated forms of CFTR, ∆264 and ∆27-264 CFTR, who can restore the function of F508-del by transcomplementation. We addressed the aims of the study using a combination of confocal microscopy and short circuit currents measurements. For the latter, CF bronchial epithelial cells (CFBE) were grown on permeable supports., Results: We show that both F508del and the truncation mutants colocalize in the ER and that both the rescued F508-del and the transcomplementing mutants reach the plasma membrane together. There was significant fluorescence resonance energy transfer (FRET) between F508-del and the transcomplementing mutants within the endoplasmic reticulum (ER), suggesting that transcomplementation occurs through a bimolecular interaction. We found that transcomplementation could increase the Isc in CFBE41o- cells stably expressing additional wt-CFTR or F508-del and in parental CFBE41o- cells expressing endogenous levels of F508-del., Conclusion: We conclude that the functional rescue of F508-del by transcomplementation occurs via a bimolecular interaction that most likely begins in the ER and continues at the plasma membrane. These results come at an opportune time for developing a gene therapy for CF and offer new treatment options for a wide range of CF patients., Competing Interests: None of the authors has any financial interests that pertain directly to this work., (© Copyright by the Author(s). Published by Cell Physiol Biochem Press.)- Published
- 2019
- Full Text
- View/download PDF
27. Rescue of CFTR NBD2 mutants N1303K and S1235R is influenced by the functioning of the autophagosome.
- Author
-
Liu Q, Sabirzhanova I, Yanda MK, Bergbower EAS, Boinot C, Guggino WB, and Cebotaru L
- Subjects
- Animals, Autophagy, Biological Transport, Blotting, Western, Cell Line, Electric Conductivity, Leucine pharmacology, Mutant Proteins drug effects, Mutant Proteins genetics, Mutation, Small Molecule Libraries pharmacology, Aminopyridines pharmacology, Autophagosomes physiology, Benzodioxoles pharmacology, Cystic Fibrosis drug therapy, Cystic Fibrosis genetics, Cystic Fibrosis Transmembrane Conductance Regulator drug effects, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Leucine analogs & derivatives
- Abstract
The missing phenylalanine at position 508, located in nucleotide-binding domain (NBD1) of the cystic fibrosis transmembrane regulator (CFTR), is the most common cystic fibrosis mutation. Severe disease-causing mutations also occur in NBD2. To provide information on potential therapeutic strategies for mutations in NBD2, we used a combination of biochemical, cell biological and electrophysiological approaches and newly created cell lines to study two disease-causing NBD2 mutants, N1303K and S1235R. We observed that neither was sensitive to E64, a cysteine protease inhibitor. However, further investigation showed that when treated with a combination of correctors, C4 + C18, both mutants also responded to E64. Further exploration to assess aggresome throughput using the autophagy regulator LC3 as a marker showed that, in the absence of correctors, N1303K showed a stalled throughput of LC3-II to the aggresome. The throughput became active again after treatment with the corrector combination C4 + C18. Confocal microscopic studies showed that the N1303K and S1235R mutant proteins both co-localized with LC3, but this co-localization was abolished by the corrector combination and, to a lesser extent, by VX-809. Both the corrector combination and VX-809 increased the CFTR chloride channel function of both mutants. We conclude that correctors have a dual effect, particularly on N1303K: they improve trafficking and function at the plasma membrane and reduce the association with autophagosomes. After treatment with correctors persistent degradation by the autophagosome may limit restoration of function. Thus, mutations in NBD2 of CFTR, in contrast to ΔF508-CFTR, may require additional personalized strategies to rescue them., (Copyright © 2018 European Cystic Fibrosis Society. Published by Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
28. A potential strategy for reducing cysts in autosomal dominant polycystic kidney disease with a CFTR corrector.
- Author
-
Yanda MK, Liu Q, and Cebotaru L
- Subjects
- Animals, Calcium metabolism, Cell Line, Cell Proliferation drug effects, Cyclic AMP metabolism, Cysts metabolism, Cysts pathology, Heat-Shock Proteins metabolism, Kidney metabolism, Kidney pathology, Mice, Mice, Inbred C57BL, Polycystic Kidney, Autosomal Dominant metabolism, Polycystic Kidney, Autosomal Dominant pathology, Transcription Factor CHOP metabolism, Aminopyridines therapeutic use, Benzodioxoles therapeutic use, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Cysts drug therapy, Kidney drug effects, Polycystic Kidney, Autosomal Dominant drug therapy
- Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is associated with progressive enlargement of cysts, leading to a decline in function and renal failure that cannot be prevented by current treatments. Mutations in pkd1 and pkd2 , encoding the polycystin 1 and 2 proteins, induce growth-related pathways, including heat shock proteins, as occurs in some cancers, raising the prospect that pharmacological interventions that target these pathways might alleviate or prevent ADPKD. Here, we demonstrate a role for VX-809, a corrector of cystic fibrosis transmembrane conductance regulator (CFTR), conventionally used to manage cystic fibrosis in reducing renal cyst growth. VX-809 reduced cyst growth in Pkd1 -knockout mice and in proximal, tubule-derived, cultured Pkd1 knockout cells. VX-809 reduced both basal and forskolin-activated cAMP levels and also decreased the expression of the adenylyl cyclase AC3 but not of AC6. VX-809 also decreased resting levels of intracellular Ca
2+ but did not affect ATP-stimulated Ca2+ release. Notably, VX-809 dramatically decreased thapsigargin-induced release of Ca2+ from the endoplasmic reticulum (ER). VX-809 also reduced the levels of heat shock proteins Hsp27, Hsp70, and Hsp90 in mice cystic kidneys, consistent with the restoration of cellular proteostasis. Moreover, VX-809 strongly decreased an ER stress marker, the GADD153 protein, and cell proliferation but had only a small effect on apoptosis. Given that administration of VX-809 is safe, this drug potentially offers a new way to treat patients with ADPKD., (© 2018 Yanda et al.)- Published
- 2018
- Full Text
- View/download PDF
29. Translational research to enable personalized treatment of cystic fibrosis.
- Author
-
Hagemeijer MC, Siegwart DJ, Strug LJ, Cebotaru L, Torres MJ, Sofoluwe A, and Beekman JM
- Subjects
- Genetic Therapy methods, Humans, Mutation, Cystic Fibrosis genetics, Cystic Fibrosis therapy, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Precision Medicine methods, Translational Research, Biomedical
- Abstract
Translational research efforts in cystic fibrosis (CF) aim to develop therapies for all subjects with CF. To reach this goal new therapies need to be developed that target multiple aspects of the disease. To enable individuals to benefit maximally from these treatments will require improved methods to tailor these therapies specifically to individuals who suffer from CF. This report highlights current examples of translational CF research efforts to reach this goal. The use of intestinal organoids and genetics to better understand individual assessment of CFTR modulator treatment effects to ultimately enable a better personalized treatment for CF subjects will be discussed. In addition, development of viral vectors and non-viral synthetic nanoparticles for delivery of mRNA, sgRNA and DNA will be highlighted. New approaches to restore function of CFTR with early premature termination codons using nanoparticle delivery of suppressor tRNAs and new insights into mechanisms of airway epithelial repair will be reviewed as well. The state-of-the-art approaches that are discussed in this review demonstrate significant progress towards the development of optimal individual therapies for CF patients, but also reveal that remaining challenges still lie ahead., (Copyright © 2017 European Cystic Fibrosis Society. Published by Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
30. Syntaxin 8 and the Endoplasmic Reticulum Processing of ΔF508-CFTR.
- Author
-
Sabirzhanova I, Boinot C, Guggino WB, and Cebotaru L
- Subjects
- Cell Line, Cystic Fibrosis genetics, Cystic Fibrosis pathology, Cystic Fibrosis Transmembrane Conductance Regulator analysis, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Endoplasmic Reticulum genetics, Endoplasmic Reticulum pathology, Gene Silencing, Humans, Protein Transport, Proteolysis, Qa-SNARE Proteins analysis, Qa-SNARE Proteins genetics, Cystic Fibrosis metabolism, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Endoplasmic Reticulum metabolism, Qa-SNARE Proteins metabolism
- Abstract
Background/aims: Cystic fibrosis (CF) is a lethal recessive disorder caused by mutations in the CF transmembrane conductance regulator (CFTR). ΔF508, the most common mutation, is a misfolded protein that is retained in the endoplasmic reticulum and degraded, precluding delivery to the cell surface [
1 ]., Methods: Here we use a combination of western blotting, immunoprecipitation, and short circuit current techniques combined with confocal microscopy to address whether the SNARE attachment protein, STX8 plays a role in ΔF508's processing and movement out of the ER., Results: Although the SNARE protein STX8 is thought to be functionally related and primarily localized to early endosomes, we show that silencing of STX8, particularly in the presence of the Vertex corrector molecule C18, rescues ΔF508-CFTR, allowing it to reach the cell surface and increasing CFTR-dependent chloride currents by approximately 2.5-fold over control values. STX8 silencing reduced the binding of quality control protein, Hsp 27, a protein that targets ΔF508-CFTR for sumoylation and subsequent degradation, to ΔF508-CFTR. STX8 silencing increased the levels of Hsp 60 a protein involving in early events in protein folding., Conclusion: STX8 knockdown creates an environment favorable for mature ΔF508 to reach the cell surface. The data also suggest that when present at normal levels, STX8 functions as part of the cell's quality control mechanism., (© 2018 The Author(s). Published by S. Karger AG, Basel.)- Published
- 2018
- Full Text
- View/download PDF
31. The CFTR-Associated Ligand Arrests the Trafficking of the Mutant ΔF508 CFTR Channel in the ER Contributing to Cystic Fibrosis.
- Author
-
Bergbower E, Boinot C, Sabirzhanova I, Guggino W, and Cebotaru L
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, COS Cells, Carrier Proteins antagonists & inhibitors, Carrier Proteins genetics, Cell Line, Cell Membrane metabolism, Chlorocebus aethiops, Cystic Fibrosis metabolism, Cystic Fibrosis Transmembrane Conductance Regulator chemistry, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Endoplasmic Reticulum metabolism, Golgi Matrix Proteins, HSP40 Heat-Shock Proteins metabolism, HSP70 Heat-Shock Proteins metabolism, HSP90 Heat-Shock Proteins metabolism, Humans, Macrolides pharmacology, Membrane Proteins antagonists & inhibitors, Membrane Proteins genetics, Membrane Transport Proteins, Phosphoproteins metabolism, Protein Binding, Protein Transport drug effects, RNA Interference, RNA, Ribosomal metabolism, RNA, Small Interfering metabolism, Sodium-Hydrogen Exchangers metabolism, Carrier Proteins metabolism, Cystic Fibrosis pathology, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Membrane Proteins metabolism
- Abstract
Background/aims: The CFTR-Associated Ligand (CAL), a PDZ domain containing protein with two coiled-coil domains, reduces cell surface WT CFTR through degradation in the lysosome by a well-characterized mechanism. However, CAL's regulatory effect on ΔF508 CFTR has remained almost entirely uninvestigated., Methods: In this study, we describe a previously unknown pathway for CAL by which it regulates the membrane expression of ΔF508 CFTR through arrest of ΔF508 CFTR trafficking in the endoplasmic reticulum (ER) using a combination of cell biology, biochemistry and electrophysiology., Results: We demonstrate that CAL is an ER localized protein that binds to ΔF508 CFTR and is degraded in the 26S proteasome. When CAL is inhibited, ΔF508 CFTR retention in the ER decreases and cell surface expression of mature functional ΔF508 CFTR is observed alongside of enhanced expression of plasma membrane scaffolding protein NHERF1. Chaperone proteins regulate this novel process, and ΔF508 CFTR binding to HSP40, HSP90, HSP70, VCP, and Aha1 changes to improve ΔF508 CFTR cell surface trafficking., Conclusion: Our results reveal a pathway in which CAL regulates the cell surface availability and intracellular retention of ΔF508 CFTR., (© 2018 The Author(s). Published by S. Karger AG, Basel.)
- Published
- 2018
- Full Text
- View/download PDF
32. Histone deacetylase 6 inhibition reduces cysts by decreasing cAMP and Ca 2+ in knock-out mouse models of polycystic kidney disease.
- Author
-
Yanda MK, Liu Q, Cebotaru V, Guggino WB, and Cebotaru L
- Subjects
- Animals, Calcium Chelating Agents pharmacology, Calcium Signaling genetics, Cell Line, Cyclic AMP genetics, Cysts genetics, Cysts pathology, Disease Models, Animal, Histone Deacetylase 6, Histone Deacetylases genetics, Mice, Mice, Knockout, Mice, Transgenic, Polycystic Kidney, Autosomal Dominant genetics, Polycystic Kidney, Autosomal Dominant pathology, Protein Kinase C genetics, Protein Kinase C metabolism, TRPP Cation Channels genetics, TRPP Cation Channels metabolism, Thapsigargin pharmacology, Calcium metabolism, Calcium Signaling drug effects, Cyclic AMP metabolism, Cysts enzymology, Histone Deacetylase Inhibitors pharmacokinetics, Histone Deacetylases metabolism, Polycystic Kidney, Autosomal Dominant enzymology
- Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is associated with progressive enlargement of multiple renal cysts, often leading to renal failure that cannot be prevented by a current treatment. Two proteins encoded by two genes are associated with ADPKD: PC1 ( pkd1 ), primarily a signaling molecule, and PC2 ( pkd2 ), a Ca
2+ channel. Dysregulation of cAMP signaling is central to ADPKD, but the molecular mechanism is unresolved. Here, we studied the role of histone deacetylase 6 (HDAC6) in regulating cyst growth to test the possibility that inhibiting HDAC6 might help manage ADPKD. Chemical inhibition of HDAC6 reduced cyst growth in PC1-knock-out mice. In proximal tubule-derived, PC1-knock-out cells, adenylyl cyclase 6 and 3 (AC6 and -3) are both expressed. AC6 protein expression was higher in cells lacking PC1, compared with control cells containing PC1. Intracellular Ca2+ was higher in PC1-knock-out cells than in control cells. HDAC inhibition caused a drop in intracellular Ca2+ and increased ATP-simulated Ca2+ release. HDAC6 inhibition reduced the release of Ca2+ from the endoplasmic reticulum induced by thapsigargin, an inhibitor of endoplasmic reticulum Ca2+ -ATPase. HDAC6 inhibition and treatment of cells with the intracellular Ca2+ chelator 1,2-bis(2-aminophenoxy)ethane- N , N , N ', N '-tetraacetic acid tetrakis(acetoxymethyl ester) reduced cAMP levels in PC1-knock-out cells. Finally, the calmodulin inhibitors W-7 and W-13 reduced cAMP levels, and W-7 reduced cyst growth, suggesting that AC3 is involved in cyst growth regulated by HDAC6. We conclude that HDAC6 inhibition reduces cell growth primarily by reducing intracellular cAMP and Ca2+ levels. Our results provide potential therapeutic targets that may be useful as treatments for ADPKD., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2017
- Full Text
- View/download PDF
33. An inhibitor of histone deacetylase 6 activity, ACY-1215, reduces cAMP and cyst growth in polycystic kidney disease.
- Author
-
Yanda MK, Liu Q, and Cebotaru L
- Subjects
- Adenylyl Cyclases metabolism, Animals, Cyclic AMP metabolism, Drug Evaluation, Preclinical, Female, Histone Deacetylase 6, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylases metabolism, Hydroxamic Acids pharmacology, Male, Mice, Inbred C57BL, Pyrimidines pharmacology, Tubulin metabolism, Histone Deacetylase Inhibitors therapeutic use, Hydroxamic Acids therapeutic use, Polycystic Kidney, Autosomal Dominant drug therapy, Pyrimidines therapeutic use
- Abstract
Adult-onset autosomal-dominant polycystic kidney disease (ADPKD) is caused by mutations in either the PKD1 or PKD2 gene, leading to malfunction of their gene products, polycystin 1 or 2. Histone deacetylase 6 (HDAC6) expression and activity are increased in PKD1 mutant renal epithelial cells. Here we studied the effect of ACY-1215, a specific HDAC6 inhibitor, on cyst growth in ADPKD. Treatment with ACY-1215 slowed cyst growth in a mouse model of ADPKD that forms massive cysts within 3 wk after knockout of polycystin 1 function. It also prevented cyst formation in MDCK.2 cells, an in vitro model of cystogenesis, and in an ADPKD cell line derived from the proximal tubules from a pkd1
-/-. mouse (PN cells). In PN cells ACY-1215 also reduced the size of already established cysts. We found that ACY-1215 lowered cAMP levels and protein expression of adenylyl cyclase 6. Our results suggest that HDAC6 could potentially serve as a therapeutic target in ADPKD., (Copyright © 2017 the American Physiological Society.)- Published
- 2017
- Full Text
- View/download PDF
34. Combination of Correctors Rescues CFTR Transmembrane-Domain Mutants by Mitigating their Interactions with Proteostasis.
- Author
-
Lopes-Pacheco M, Boinot C, Sabirzhanova I, Rapino D, and Cebotaru L
- Subjects
- Anilides pharmacology, Animals, COS Cells, Chlorocebus aethiops, Cysteine Proteinase Inhibitors pharmacology, Cystic Fibrosis Transmembrane Conductance Regulator chemistry, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Endoplasmic Reticulum metabolism, HEK293 Cells, Humans, Hydroxamic Acids pharmacology, Immunoprecipitation, Leupeptins pharmacology, Mutagenesis, Site-Directed, Proteasome Endopeptidase Complex chemistry, Protein Binding, Protein Stability, Proteolysis drug effects, Temperature, Transfection, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Proteasome Endopeptidase Complex metabolism
- Abstract
Background/aims: Premature degradation of mutated cystic fibrosis transmembrane conductance regulator (CFTR) protein causes cystic fibrosis (CF), the commonest Mendelian disease in Caucasians. Despite recent advances in precision medicines for CF patients, many CFTR mutants have not been characterized and the effects of these new therapeutic approaches are still unclear for those mutants., Methods: Cells transfected or stably expressing four CFTR transmembrane-domain mutants (G85E, E92K, L1077P, and M1101K) were used to: 1) characterize the mutants according to their protein expression, thermal sensitivity, and degradation pathways; 2) evaluate the effects of correctors in rescuing them; and 3) explore the effects of correctors on CFTR interactions with proteostasis components., Results: All four mutants exhibited lower protein expression than did wild type-CFTR, and they were degraded by proteasomes and aggresomes. At low temperature, only cells expressing the mutants L1077P and M1101K exhibited increased CFTR maturation. Co-administration of C4 and C18 showed the greatest effect, restoring functional expression and partial stability of CFTR bearing E92K, L1077P, or M1101K at the cell surface. However, this treatment was inefficient in rectifying the defect of CFTR bearing G85E. Correctors rescued CFTR mutants by reducing their interactions with proteostasis components associated with protein retention in the endoplasmic reticulum and ubiquitination., Conclusion: Co-administration of C4 and C18 rescued CFTR transmembrane-domain mutants by remodeling the CFTR interactome., (© 2017 The Author(s)Published by S. Karger AG, Basel.)
- Published
- 2017
- Full Text
- View/download PDF
35. Inhibition of histone deacetylase 6 activity reduces cyst growth in polycystic kidney disease.
- Author
-
Cebotaru L, Liu Q, Yanda MK, Boinot C, Outeda P, Huso DL, Watnick T, Guggino WB, and Cebotaru V
- Subjects
- Animals, Cell Proliferation drug effects, Chlorides blood, Chlorides metabolism, Cyclic AMP blood, Disease Models, Animal, Dogs, Down-Regulation, Epithelial Cells metabolism, Female, Histone Deacetylase 6, Histone Deacetylases genetics, Humans, Kidney enzymology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Polycystic Kidney, Autosomal Dominant genetics, TRPP Cation Channels genetics, Anilides pharmacology, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Epithelial Cells physiology, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylases metabolism, Hydroxamic Acids pharmacology, Kidney drug effects, Polycystic Kidney, Autosomal Dominant metabolism
- Abstract
Abnormal proliferation of cyst-lining epithelium and increased intracystic fluid secretion via the cystic fibrosis transmembrane conductance regulator (CFTR) are thought to contribute to cyst growth in autosomal dominant polycystic kidney disease (ADPKD). Histone deacetylase 6 (HDAC6) expression and activity are increased in certain cancers, neurodegenerative diseases, and in Pkd1-mutant renal epithelial cells. Inhibition of HDAC6 activity with specific inhibitors slows cancer growth. Here we studied the effect of tubacin, a specific HDAC6 inhibitor, on cyst growth in polycystic kidney disease. Treatment with tubacin prevented cyst formation in MDCK cells, an in vitro model of cystogenesis. Cyclic AMP stimulates cell proliferation and activates intracystic CFTR-mediated chloride secretion in ADPKD. Treatment with tubacin downregulated cyclic AMP levels, inhibited cell proliferation, and inhibited cyclic AMP-activated CFTR chloride currents in MDCK cells. We also found that tubacin reduced cyst growth by inhibiting proliferation of cyst-lining epithelial cells, downregulated cyclic AMP levels, and improved renal function in a Pkd1-conditional mouse model of ADPKD. Thus, HDAC6 could play a role in cyst formation and could serve as a potential therapeutic target in ADPKD., (Copyright © 2016 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
36. CFTR Controls the Activity of NF-κB by Enhancing the Degradation of TRADD.
- Author
-
Wang H, Cebotaru L, Lee HW, Yang Q, Pollard BS, Pollard HB, and Guggino WB
- Subjects
- Adaptor Proteins, Signal Transducing, Carrier Proteins metabolism, Cell Line, Cell Movement drug effects, Golgi Matrix Proteins, HEK293 Cells, Humans, Membrane Proteins metabolism, Membrane Transport Proteins, Protein Binding drug effects, Time Factors, Tumor Necrosis Factor-alpha pharmacology, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, NF-kappa B metabolism, Proteolysis drug effects, TNF Receptor-Associated Death Domain Protein metabolism
- Abstract
Background/aims: Chronic lung infection in cystic fibrosis leads to an inflammatory response that persists because of the chronic presence of bacteria and ultimately leads to a catastrophic failure of lung function., Methods: We use a combination of biochemistry, cell and molecular biology to study the interaction of TRADD, a key adaptor molecule in TNFα signaling, with CFTR in the regulation of NFκB., Results: We show that Wt CFTR binds to and colocalizes with TRADD. TRADD is a key signaling intermediate connecting TNFα with activation of NFκB. By contrast, ΔF508 CFTR does not bind to TRADD. NF-κB activation is higher in CFBE expressing ΔF508 CFTR than in cells expressing Wt CFTR. However, this differential effect is abolished when TRADD levels are knocked down. Transfecting Wt CFTR into CFBE cells reduces NF-κB activity. However the reduction is abolished by the CFTR chloride transport inhibitor-172. Consistently, transfecting in the correctly trafficked CFTR conduction mutants G551D or S341A also fail to reduce NFκB activity. Thus CFTR must be functional if it is to regulate NF-κB activity. We also found that TNFα produced a greater increase in NF-κB activity in CFBE cells than in the same cell when Wt CFTR-corrected. Consistently, the effect is also abolished when TRADD is knocked down by shRNA. Thus, Wt CFTR control of TRADD modulates the physiological activation of NF-κB by TNFα. Based on studies with proteosomal and lysosomal inhibitors, the mechanism by which Wt CFTR, but not ΔF508 CFTR, suppresses TRADD is by lysosomal degradation., Conclusion: We have uncovered a novel mechanism whereby Wt CFTR regulates TNFα signaling by enhancing TRADD degradation. Thus by reducing the levels of TRADD, Wt CFTR suppresses downstream proinflammatory NFκB signaling. By contrast, suppression of NF-κB activation fails in CF cells expressing ΔF508 CFTR., (© 2016 The Author(s) Published by S. Karger AG, Basel.)
- Published
- 2016
- Full Text
- View/download PDF
37. Regulation of CFTR Expression and Arginine Vasopressin Activity Are Dependent on Polycystin-1 in Kidney-Derived Cells.
- Author
-
de Lemos Barbosa CM, Souza-Menezes J, Amaral AG, Onuchic LF, Cebotaru L, Guggino WB, and Morales MM
- Subjects
- Animals, Antidiuretic Agents pharmacology, Cell Line, Cyclic AMP metabolism, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Epithelial Cells cytology, Epithelial Cells drug effects, Epithelial Cells metabolism, Kidney cytology, Mice, Mice, Inbred CFTR, Polycystic Kidney, Autosomal Dominant genetics, Polycystic Kidney, Autosomal Dominant pathology, RNA, Messenger metabolism, Real-Time Polymerase Chain Reaction, TRPP Cation Channels genetics, Transfection, Arginine Vasopressin pharmacology, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Gene Expression Regulation drug effects, TRPP Cation Channels metabolism
- Abstract
Background: Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the development of multiple, progressive, fluid-filled renal cysts that distort the renal parenchyma, leading to end-stage renal failure, mainly after the fifth decade of life. ADPKD is caused by a mutation in the PKD1 or PKD2 genes that encode polycystin-1 (PC-1) and polycystin-2 (PC-2), respectively. PC-1 is an important regulator of several signaling pathways and PC-2 is a nonselective calcium channel. The CFTR chloride channel is responsible for driving net fluid secretion into the cysts, promoting cyst growth. Arginine vasopressin hormone (AVP), in turn, is capable of increasing cystic intracellular cAMP, contributing to cell proliferation, transepithelial fluid secretion, and therefore to disease progression. The aim of this study was to assess if AVP can modulate CFTR and whether PC-1 plays a role in this potential modulation., Methods: M1 cells, derived from mouse cortical collecting duct, were used in the current work. The cells were treated with 10-7 M AVP hormone and divided into two main groups: transfected cells superexpressing PC-1 (Transf) and cells not transfected (Ctrl). CFTR expression was assessed by immunodetection, CFTR mRNA levels were quantified by quantitative reverse transcription-polymerase chain reaction, and CFTR net ion transport was measured using the Ussing chamber technique., Results: AVP treatment increased the levels of CFTR protein and mRNA. CFTR short-circuit currents were also increased. However, when PC-1 was overexpressed in M1 cells, no increase in any of these parameters was detected., Conclusions: CFTR chloride channel expression is increased by AVP in M1 cells and PC-1 is capable of regulating this modulation., (© 2016 S. Karger AG, Basel.)
- Published
- 2016
- Full Text
- View/download PDF
38. STIM1fl/fl Ksp-Cre Mouse has Impaired Renal Water Balance.
- Author
-
Cebotaru L, Cebotaru V, Wang H, Arend LJ, and Guggino WB
- Subjects
- Animals, Calcium blood, Calcium urine, Creatinine blood, Creatinine urine, Dietary Proteins administration & dosage, Dietary Proteins pharmacology, Kidney Concentrating Ability genetics, Kinesins genetics, Mice, Knockout, Mice, Transgenic, Renal Insufficiency genetics, Renal Insufficiency metabolism, Stromal Interaction Molecule 1 genetics, Urea blood, Urea urine, Urination drug effects, Water-Electrolyte Balance genetics, Kidney metabolism, Kinesins metabolism, Stromal Interaction Molecule 1 metabolism, Water metabolism
- Abstract
Background/aim: STIM1 is as an essential component in store operated Ca2+ entry. However give the paucity of information on the role of STIM1 in kidney, the aim was to study the function of STIM1 in the medulla of the kidney., Methods: we crossed a Ksp-cre mouse with another mouse containing two loxP sites flanking Exon 6 of STIM1. The Ksp-cre mouse is based upon the Ksp-cadherin gene promoter which expresses cre recombinase in developing nephrons, collecting ducts (SD) and thick ascending limbs (TAL) of the loop of Henle., Results: The offspring of these mice are viable without gross morphological changes, however, we noticed that the STIM1 Ksp-cre knockout mice produced more urine compared to control. To examine this more carefully, we fed mice low (LP) and high protein (HP) diets respectively. When mice were fed HP diet STIM1 ko mice had significantly increased urinary volume and lower specific gravity compared to wt mice. In STIM1 ko mice fed HP diet urine creatinine and urea were significantly lower compared to wt mice fed HP diet, however the fractional excretion was the same., Conclusion: These data support the idea that STIM1 ko mice have impaired urinary concentrating ability when challenged with HP diet is most likely caused by impaired Ca2+-dependent signal transduction through the vasopressin receptor cascade., (© 2016 The Author(s) Published by S. Karger AG, Basel.)
- Published
- 2016
- Full Text
- View/download PDF
39. Combination of Correctors Rescue ΔF508-CFTR by Reducing Its Association with Hsp40 and Hsp27.
- Author
-
Lopes-Pacheco M, Boinot C, Sabirzhanova I, Morales MM, Guggino WB, and Cebotaru L
- Subjects
- Animals, COS Cells, Chlorocebus aethiops, Cystic Fibrosis Transmembrane Conductance Regulator genetics, HEK293 Cells, Humans, Mutation, Protein Binding, Temperature, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, HSP27 Heat-Shock Proteins metabolism, HSP40 Heat-Shock Proteins metabolism
- Abstract
Correcting the processing of ΔF508-CFTR, the most common mutation in cystic fibrosis, is the major goal in the development of new therapies for this disease. Here, we determined whether ΔF508 could be rescued by a combination of small-molecule correctors, and identified the mechanism by which correctors rescue the trafficking mutant of cystic fibrosis transmembrane conductance regulator (CFTR). We transfected COS-7 cells with ΔF508, created HEK-293 stably expressing ΔF508, and utilized CFBE41o(-) cell lines stably transduced with ΔF508. As shown previously, ΔF508 expressed less protein, was unstable at physiological temperature, and rapidly degraded. When the cells were treated with the combination C18 + C4 the mature C-band was expressed at the cell surface. After treatment with C18 + C4, we saw a lower rate of protein disappearance after translation was stopped with cycloheximide. To understand how this rescue occurs, we evaluated the change in the binding of proteins involved in endoplasmic reticulum-associated degradation, such as Hsp27 (HspB1) and Hsp40 (DnaJ). We saw a dramatic reduction in binding to heat shock proteins 27 and 40 following combined corrector therapy. siRNA experiments confirmed that a reduction in Hsp27 or Hsp40 rescued CFTR in the ΔF508 mutant, but the rescue was not additive or synergistic with C4 + 18 treatment, indicating these correctors shared a common pathway for rescue involving a network of endoplasmic reticulum-associated degradation proteins., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
40. Rescuing Trafficking Mutants of the ATP-binding Cassette Protein, ABCA4, with Small Molecule Correctors as a Treatment for Stargardt Eye Disease.
- Author
-
Sabirzhanova I, Lopes Pacheco M, Rapino D, Grover R, Handa JT, Guggino WB, and Cebotaru L
- Subjects
- ATP-Binding Cassette Transporters chemistry, ATP-Binding Cassette Transporters genetics, Amino Acid Sequence, Anilides pharmacology, Cystic Fibrosis Transmembrane Conductance Regulator chemistry, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Enzyme Inhibitors pharmacology, Gene Expression, HEK293 Cells, HSP27 Heat-Shock Proteins genetics, Histone Deacetylase 6, Histone Deacetylases genetics, Humans, Hydroxamic Acids pharmacology, Macrolides pharmacology, Macular Degeneration congenital, Macular Degeneration drug therapy, Macular Degeneration genetics, Macular Degeneration metabolism, Molecular Sequence Data, Mutation, Protein Transport, Proteolysis, Sequence Homology, Amino Acid, Signal Transduction, Stargardt Disease, Transgenes, ATP-Binding Cassette Transporters metabolism, Aminopyridines pharmacology, Benzodioxoles pharmacology, HSP27 Heat-Shock Proteins metabolism, Histone Deacetylases metabolism, Protective Agents pharmacology
- Abstract
Stargardt disease is the most common form of early onset macular degeneration. Mutations in ABCA4, a member of the ATP-binding cassette (ABC) family, are associated with Stargardt disease. Here, we have examined two disease-causing mutations in the NBD1 region of ABCA4, R1108C, and R1129C, which occur within regions of high similarity with CFTR, another ABC transporter gene, which is associated with cystic fibrosis. We show that R1108C and R1129C are both temperature-sensitive processing mutants that engage the cellular quality control mechanism and show a strong interaction with the chaperone Hsp 27. Both mutant proteins also interact with HDCAC6 and are degraded in the aggresome. We also demonstrate that novel corrector compounds that are being tested as treatment for cystic fibrosis, such as VX-809, can rescue the processing of the ABCA4 mutants, particularly their expression at the cell surface, and can reduce their binding to HDAC6. Thus, our data suggest that VX-809 can potentially be developed as a new therapy for Stargardt disease, for which there is currently no treatment., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
41. Rescue of NBD2 mutants N1303K and S1235R of CFTR by small-molecule correctors and transcomplementation.
- Author
-
Rapino D, Sabirzhanova I, Lopes-Pacheco M, Grover R, Guggino WB, and Cebotaru L
- Subjects
- Animals, Biological Transport, COS Cells, Chlorocebus aethiops, Cystic Fibrosis pathology, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, HEK293 Cells, Humans, Protein Binding, Cystic Fibrosis drug therapy, Cystic Fibrosis genetics, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Genetic Complementation Test, Mutation genetics, Small Molecule Libraries pharmacology
- Abstract
Although, the most common Cystic Fibrosis mutation, ΔF508, in the cystic fibrosis transmembrane regulator. (CFTR), is located in nucleotide binding domain (NBD1), disease-causing mutations also occur in NBD2. To provide information on potential therapeutic strategies for mutations in NBD2, we studied, using a combination of biochemical approaches and newly created cell lines, two disease-causing NBD2 mutants, N1303K and S1235R. Surprisingly, neither was rescued by low temperature. Inhibition of proteasomes with MG132 or aggresomes with tubacin rescued the immature B and mature C bands of N1303K and S1235R, indicating that degradation occurs via proteasomes and aggresomes. We found no effect of the lysosome inhibitor E64. Thus, our results show that these NBD2 mutants are processing mutants with unique characteristics. Several known correctors developed to rescue ΔF508-CFTR, when applied either alone or in combination, significantly increased the maturation of bands B and C of both NBD 2 mutants. The best correction occurred with the combinations of C4 plus C18 or C3 plus C4. Co-transfection of truncated CFTR (∆27-264) into stably transfected cells was also able to rescue them. This demonstrates for the first time that transcomplementation with a truncated version of CFTR can rescue NBD2 mutants. Our results show that the N1303K mutation has a more profound effect on NBD2 processing than S1235R and that small-molecule correctors increase the maturation of bands B and C in NBD2 mutants. In addition, ∆27-264 was able to transcomplement both NDB2 mutants. We conclude that differences and similarities occur in the impact of mutations on NBD2 when compared to ΔF508-CFTR suggesting that individualized strategies may be needed to restore their function. Finally our results are important because they suggest that gene or corrector molecule therapies either alone or in combination individualized for NBD2 mutants may be beneficial for patients bearing N1303K or S1235R mutations.
- Published
- 2015
- Full Text
- View/download PDF
42. Polycystin-1 negatively regulates Polycystin-2 expression via the aggresome/autophagosome pathway.
- Author
-
Cebotaru V, Cebotaru L, Kim H, Chiaravalli M, Boletta A, Qian F, and Guggino WB
- Subjects
- Animals, Dogs, Down-Regulation, Histone Deacetylase 6, Histone Deacetylases metabolism, Kidney metabolism, Madin Darby Canine Kidney Cells, Metabolic Networks and Pathways, Mice, Polycystic Kidney, Autosomal Dominant genetics, Polycystic Kidney, Autosomal Dominant metabolism, TRPP Cation Channels antagonists & inhibitors, TRPP Cation Channels genetics, Autophagy, Phagosomes, TRPP Cation Channels metabolism
- Abstract
Mutations of the PKD1 and PKD2 genes, encoding polycystin-1 (PC1) and polycystin-2 (PC2), respectively, lead to autosomal dominant polycystic kidney disease. Interestingly, up-regulation or down-regulation of PKD1 or PKD2 leads to polycystic kidney disease in animal models, but their interrelations are not completely understood. We show here that full-length PC1 that interacts with PC2 via a C-terminal coiled-coil domain regulates PC2 expression in vivo and in vitro by down-regulating PC2 expression in a dose-dependent manner. Expression of the pathogenic mutant R4227X, which lacks the C-terminal coiled-coil domain, failed to down-regulate PC2 expression, suggesting that PC1-PC2 interaction is necessary for PC2 regulation. The proteasome and autophagy are two pathways that control protein degradation. Proteins that are not degraded by proteasomes precipitate in the cytoplasm and are transported via histone deacetylase 6 (HDAC6) toward the aggresomes. We found that HDAC6 binds to PC2 and that expression of full-length PC1 accelerates the transport of the HDAC6-PC2 complex toward aggresomes, whereas expression of the R4227X mutant fails to do so. Aggresomes are engulfed by autophagosomes, which then fuse with the lysosome for degradation; this process is also known as autophagy. We have now shown that PC1 overexpression leads to increased degradation of PC2 via autophagy. Interestingly, PC1 does not activate autophagy generally. Thus, we have now uncovered a new pathway suggesting that when PC1 is expressed, PC2 that is not bound to PC1 is directed to aggresomes and subsequently degraded via autophagy, a control mechanism that may play a role in autosomal dominant polycystic kidney disease pathogenesis.
- Published
- 2014
- Full Text
- View/download PDF
43. Complement yourself: Transcomplementation rescues partially folded mutant proteins.
- Author
-
Cebotaru L and Guggino WB
- Abstract
Cystic Fibrosis (CF) is an autosomal disease associated with malfunction in fluid and electrolyte transport across several mucosal membranes. The most common mutation in CF is an in-frame three-base pair deletion that removes a phenylalanine at position 508 in the first nucleotide-binding domain of the cystic fibrosis conductance regulator (CFTR) chloride channel. This mutation has been studied extensively and leads to biosynthetic arrest of the protein in the endoplasmic reticulum and severely reduced channel activity. This review discusses a novel method of rescuing ΔF508 with transcomplementation, which occurs when smaller fragments of CFTR containing the wild-type nucleotide binding domain are co-expressed with the ΔF508 deletion mutant. Transcomplementation rescues the processing and channel activity of ΔF508 and reduces its rate of degradation in airway epithelial cells. To apply transcomplementation as a therapy would require that the cDNA encoding the truncated CFTR be delivered to cells. We also discuss a gene therapeutic approach based on delivery of a truncated form of CFTR to airway cells using adeno-associated viral vectors.
- Published
- 2014
- Full Text
- View/download PDF
44. Correcting the cystic fibrosis disease mutant, A455E CFTR.
- Author
-
Cebotaru L, Rapino D, Cebotaru V, and Guggino WB
- Subjects
- Anilides pharmacology, Animals, Boronic Acids pharmacology, Bortezomib, COS Cells, Chlorocebus aethiops, Cycloheximide pharmacology, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Gene Expression, Genetic Complementation Test, Half-Life, Humans, Hydroxamic Acids pharmacology, Leupeptins pharmacology, Plasmids, Proteasome Endopeptidase Complex metabolism, Proteolysis drug effects, Pyrazines pharmacology, Transfection, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Mutation, Proteasome Endopeptidase Complex drug effects, Proteasome Inhibitors pharmacology, Small Molecule Libraries pharmacology
- Abstract
Cystic fibrosis is caused by more than 1000 mutations, the most common being the ΔF508 mutation. These mutations have been divided into five classes [1], with ΔF508 CFTR in class II. Here we have studied the class V mutation A455E. We report that the mature and immature bands of A455E are rapidly degraded primarily by proteasomes; the short protein half-life of this mutant therefore resembles that of ΔF508 CFTR. A455E could be rescued by treatment of the cells with proteasome inhibitors. Furthermore, co-transfection of A455E with the truncation mutant Δ264 CFTR also rescued the mature C band, indicating that A455E can be rescued by transcomplementation. We found that Δ264 CFTR bound to A455E, forming a bimolecular complex. Treatment with the compound correctors C3 and C4 also rescued A455E. These results are significant because they show that although ΔF508 belongs to a different class than A455E, it can be rescued by the same strategies, offering therapeutic promise to patients with Class V mutations.
- Published
- 2014
- Full Text
- View/download PDF
45. Transcomplementation by a truncation mutant of cystic fibrosis transmembrane conductance regulator (CFTR) enhances ΔF508 processing through a biomolecular interaction.
- Author
-
Cebotaru L, Woodward O, Cebotaru V, and Guggino WB
- Subjects
- Amino Acid Sequence, Animals, CHO Cells, COS Cells, Chlorocebus aethiops, Cricetinae, Cricetulus, Cystic Fibrosis genetics, Cystic Fibrosis metabolism, Cystic Fibrosis therapy, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Dependovirus, Genetic Complementation Test, Genetic Therapy methods, Genetic Vectors, Humans, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Mutation, Missense, Sequence Deletion
- Abstract
We previously showed that a truncation mutant of CFTR missing the first four transmembrane segments of TMD1, Δ264 CFTR, binds to key elements in the ER quality control mechanism to increase the amounts of the mature C band of both wt and ΔF508 CFTR through transcomplementation. Here, we created a new construct, Δ27-264 CFTR. Even though Δ27-264 CFTR is rapidly degraded in the proteasome, steady state protein can be detected by Western blot. Δ27-264 CFTR can also increase the amounts of the mature C band of both wt and ΔF508 CFTR through transcomplementation. Electrophysiology experiments show that Δ27-264 CFTR can restore chloride channel currents. Further experiments with the conduction mutant S341A show conclusively that currents are indeed generated by rescued channel function of ΔF508 CFTR. Immunoprecipitation studies show that Δ27-264 binds to ΔF508-CFTR, suggesting a bimolecular interaction. Thus the adeno-associated viral vector, rAAV-Δ27-264 CFTR, is a highly promising CF gene therapy vector, because it increases the amount of mature band C protein both from wt and ΔF508 CFTR, and rescues channel activity of ΔF508 CFTR.
- Published
- 2013
- Full Text
- View/download PDF
46. C1q/TNF-related protein-12 (CTRP12), a novel adipokine that improves insulin sensitivity and glycemic control in mouse models of obesity and diabetes.
- Author
-
Wei Z, Peterson JM, Lei X, Cebotaru L, Wolfgang MJ, Baldeviano GC, and Wong GW
- Subjects
- Adenoviridae, Adipocytes pathology, Adipokines genetics, Adipose Tissue pathology, Animals, Cells, Cultured, Diabetes Mellitus, Experimental genetics, Diabetes Mellitus, Experimental pathology, Disease Models, Animal, Gluconeogenesis genetics, Hepatocytes pathology, Homeostasis genetics, Humans, Insulin genetics, Insulin metabolism, Male, Mice, Mice, Inbred AKR, Mice, Inbred BALB C, Mice, Inbred DBA, Mice, Obese, Obesity genetics, Obesity pathology, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Signal Transduction genetics, Transduction, Genetic, Adipocytes metabolism, Adipokines blood, Adipose Tissue metabolism, Diabetes Mellitus, Experimental blood, Hepatocytes metabolism, Obesity blood
- Abstract
Despite the prevalence of insulin resistance and type 2 diabetes mellitus, their underlying mechanisms remain incompletely understood. Many secreted endocrine factors and the intertissue cross-talk they mediate are known to be dysregulated in type 2 diabetes mellitus. Here, we describe CTRP12, a novel adipokine with anti-diabetic actions. The mRNA and circulating levels of CTRP12 were decreased in a mouse model of obesity, but its expression in adipocytes was increased by the anti-diabetic drug rosiglitazone. A modest rise in circulating levels of CTRP12 by recombinant protein administration was sufficient to lower blood glucose in wild-type, leptin-deficient ob/ob, and diet-induced obese mice. A short term elevation of serum CTRP12 by adenovirus-mediated expression improved glucose tolerance and insulin sensitivity, normalized hyperglycemia and hyperinsulinemia, and lowered postprandial insulin resistance in obese and diabetic mice. CTRP12 improves insulin sensitivity in part by enhancing insulin signaling in the liver and adipose tissue. Further, CTRP12 also acts in an insulin-independent manner; in cultured hepatocytes and adipocytes, CTRP12 directly activated the PI3K-Akt signaling pathway to suppress gluconeogenesis and promote glucose uptake, respectively. Collectively, these data establish CTRP12 as a novel metabolic regulator linking adipose tissue to whole body glucose homeostasis through insulin-dependent and independent mechanisms.
- Published
- 2012
- Full Text
- View/download PDF
47. Dexamethasone regulates CFTR expression in Calu-3 cells with the involvement of chaperones HSP70 and HSP90.
- Author
-
Prota LF, Cebotaru L, Cheng J, Wright J, Vij N, Morales MM, and Guggino WB
- Subjects
- Amino Acid Motifs, Cell Line, Cell Membrane metabolism, Culture Media metabolism, Cycloheximide therapeutic use, Dose-Response Relationship, Drug, Glucocorticoids metabolism, Glucocorticoids pharmacology, Humans, Mifepristone pharmacology, Protein Structure, Tertiary, Real-Time Polymerase Chain Reaction methods, Time Factors, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Dexamethasone pharmacology, HSP70 Heat-Shock Proteins metabolism, HSP90 Heat-Shock Proteins metabolism
- Abstract
Background: Dexamethasone is widely used for pulmonary exacerbation in patients with cystic fibrosis, however, not much is known about the effects of glucocorticoids on the wild-type cystic fibrosis channel transmembrane regulator (CFTR). Our aim was to determine the effects of dexamethasone treatment on wild-type CFTR expression., Methods and Results: Dose-response (1 nM to 10 µM) and time course (3 to 48 h) curves were generated for dexamethasone for mRNA expression in Calu-3 cells using a real-time PCR. Within 24 h, dexamethasone (10 nM) showed a 0.3-fold decrease in CFTR mRNA expression, and a 3.2-fold increase in αENaC mRNA expression compared with control groups. Dexamethasone (10 nM) induced a 1.97-fold increase in the total protein of wild-type CFTR, confirmed by inhibition by mifepristone. To access surface protein expression, biotinylation followed by Western blotting showed that dexamethasone treatment led to a 2.35-fold increase in the amount of CFTR in the cell surface compared with the untreated control groups. Once protein translation was inhibited with cycloheximide, dexamethasone could not increase the amount of CFTR protein. Protein stability was assessed by inhibition of protein synthesis with cycloheximide (50 µg/ml) at different times in cells treated with dexamethasone and in untreated cells. Dexamethasone did not alter the degradation of wild-type CFTR. Assessment of the B band of CFTR within 15 min of metabolic pulse labeling showed a 1.5-fold increase in CFTR protein after treatment with dexamethasone for 24 h. Chaperone 90 (HSP90) binding to CFTR increased 1.55-fold after treatment with dexamethasone for 24 h, whereas chaperone 70 (HSP70) binding decreased 0.30 fold in an immunoprecipitation assay., Conclusion: Mature wild-type CFTR protein is regulated by dexamethasone post transcription, involving cotranslational mechanisms with HSP90 and HSP70, which enhances maturation and expression of wild-type CFTR.
- Published
- 2012
- Full Text
- View/download PDF
48. Enterohemorrhagic Escherichia coli infection stimulates Shiga toxin 1 macropinocytosis and transcytosis across intestinal epithelial cells.
- Author
-
Lukyanenko V, Malyukova I, Hubbard A, Delannoy M, Boedeker E, Zhu C, Cebotaru L, and Kovbasnjuk O
- Subjects
- Actins metabolism, Cell Line, Colon metabolism, Colon microbiology, Escherichia coli Infections microbiology, Humans, Intestinal Mucosa microbiology, Myosin Type II metabolism, cdc42 GTP-Binding Protein metabolism, Enterohemorrhagic Escherichia coli, Escherichia coli Infections metabolism, Intestinal Mucosa metabolism, Pinocytosis, Shiga Toxin 1 metabolism, Transcytosis
- Abstract
Gastrointestinal infection with Shiga toxins producing enterohemorrhagic Escherichia coli causes the spectrum of gastrointestinal and systemic complications, including hemorrhagic colitis and hemolytic uremic syndrome, which is fatal in ∼10% of patients. However, the molecular mechanisms of Stx endocytosis by enterocytes and the toxins cross the intestinal epithelium are largely uncharacterized. We have studied Shiga toxin 1 entry into enterohemorrhagic E. coli-infected intestinal epithelial cells and found that bacteria stimulate Shiga toxin 1 macropinocytosis through actin remodeling. This enterohemorrhagic E. coli-caused macropinocytosis occurs through a nonmuscle myosin II and cell division control 42 (Cdc42)-dependent mechanism. Macropinocytosis of Shiga toxin 1 is followed by its transcytosis to the basolateral environment, a step that is necessary for its systemic spread. Inhibition of Shiga toxin 1 macropinocytosis significantly decreases toxin uptake by intestinal epithelial cells and in this way provides an attractive, antibiotic-independent strategy for prevention of the harmful consequences of enterohemorrhagic E. coli infection.
- Published
- 2011
- Full Text
- View/download PDF
49. Syntaxin 6 and CAL mediate the degradation of the cystic fibrosis transmembrane conductance regulator.
- Author
-
Cheng J, Cebotaru V, Cebotaru L, and Guggino WB
- Subjects
- Adaptor Proteins, Signal Transducing, Cell Line, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Gene Silencing, Genes, Dominant, Golgi Matrix Proteins, Humans, Membrane Transport Proteins, Microscopy, Confocal methods, Models, Biological, Mutation, Plasmids metabolism, RNA, Small Interfering metabolism, Carrier Proteins physiology, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Gene Expression Regulation, Membrane Proteins physiology, Qa-SNARE Proteins physiology
- Abstract
The PDZ domain-containing protein CAL mediates lysosomal trafficking and degradation of CFTR. Here we demonstrate the involvement of a CAL-binding SNARE protein syntaxin 6 (STX6) in this process. Overexpression of STX6, which colocalizes and coimmunoprecipitates with CAL, dramatically reduces the steady-state level and stability of CFTR. Conversely, overexpression of a STX6 dominant-negative mutant increases CFTR. Silencing endogenous STX6 increases CFTR but has no effect on DeltaTRL-CFTR, which cannot bind to CAL. Silencing CAL eliminates the effect of STX6 on CFTR. Both results suggest a dependence of CAL on STX6 function. Consistent with its Golgi localization, STX6 does not bind to ER-localized DeltaF508-CFTR. Silencing STX6 has no effect on DeltaF508-CFTR expression. However, overexpression of STX6 coimmunoprecipitates with and reduces temperature-rescued DeltaF508-CFTR that escapes ER degradation. Conversely, silencing STX6 enhances the effect of low temperature in rescuing DeltaF508-CFTR. Finally, in human bronchial epithelial cells, silencing endogenous STX6 leads to increases in protein levels and Cl(-) currents of both wild-type and temperature-rescued CFTR. We have identified STX6 as a new component of the CAL complex that regulates the abundance and function of CFTR at the post-ER level. Our results suggest a therapeutic role of STX6 in enhancing rescued DeltaF508-CFTR.
- Published
- 2010
- Full Text
- View/download PDF
50. Dual reporter comparative indexing of rAAV pseudotyped vectors in chimpanzee airway.
- Author
-
Flotte TR, Fischer AC, Goetzmann J, Mueller C, Cebotaru L, Yan Z, Wang L, Wilson JM, Guggino WB, and Engelhardt JF
- Subjects
- Animals, Biological Evolution, Gene Transfer Techniques, Genetic Vectors, Humans, Immune System, Luciferases metabolism, Pan troglodytes, Phylogeny, Species Specificity, T-Lymphocytes metabolism, Transgenes, Dependovirus genetics, Genes, Reporter, Respiratory System metabolism
- Abstract
Selecting the most efficient recombinant adeno-associated virus (rAAV) serotype for airway gene therapy has been difficult due to cross-specific differences in tropism and immune response between humans and animal models. Chimpanzees--the closest surviving genetic relative of humans--provide a valuable opportunity to select the most effective serotypes for clinical trials in humans. However, designing informative experiments using this protected species is challenging due to limited availability and experimental regulations. We have developed a method using Renilla luciferase (RL) and firefly luciferase (FL) reporters to directly index the relative transduction and immune response of two promising rAAV serotypes following lung coinfection. Analysis of differential luciferase activity in chimpanzee airway brushings demonstrated a 20-fold higher efficiency for rAAV1 over rAAV5 at 90 days, a finding that was similar in polarized human airway epithelia. T-cell responses to AAV5 capsid were stronger than AAV1 capsid. This dual vector indexing approach may be useful in selecting lead vector serotypes for clinical gene therapy and suggests rAAV1 is preferred for cystic fibrosis.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.