1
|
Abstract
Ubiquitously expressed throughout the body, ATP-sensitive potassium (KATP) channels couple cellular metabolism to electrical activity in multiple tissues; their unique assembly as four Kir6 pore-forming subunits and four sulfonylurea receptor (SUR) subunits has resulted in a large armory of selective channel opener and inhibitor drugs. The spectrum of monogenic pathologies that result from gain- or loss-of-function mutations in these channels, and the potential for therapeutic correction of these pathologies, is now clear. However, while available drugs can be effective treatments for specific pathologies, cross-reactivity with the other Kir6 or SUR subfamily members can result in drug-induced versions of each pathology and may limit therapeutic usefulness. This review discusses the background to KATP channel physiology, pathology, and pharmacology and considers the potential for more specific or effective therapeutic agents.
Collapse
Affiliation(s)
- Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases and Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA;
| |
Collapse
|
2
|
Nagar S, Radice C, Tuohy R, Stevens R, Bennyhoff D, Korzekwa K. The Rat Continuous Intestine Model Predicts the Impact of Particle Size and Transporters on the Oral Absorption of Glyburide. Mol Pharm 2023; 20:219-231. [PMID: 36541850 DOI: 10.1021/acs.molpharmaceut.2c00597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Oral drug absorption is known to be impacted by the physicochemical properties of drugs, properties of oral formulations, and physiological characteristics of the intestine. The goal of the present study was to develop a mathematical model to predict the impact of particle size, feeding time, and intestinal transporter activity on oral absorption. A previously published rat continuous intestine absorption model was extended for solid drug absorption. The impact of active pharmaceutical ingredient particle size was evaluated with glyburide (GLY) as a model drug. Two particle size suspensions of glyburide were prepared with average particle sizes of 42.7 and 4.1 μm. Each suspension was dosed as a single oral gavage to male Sprague Dawley rats, and concentration-time (C-t) profiles of glyburide were measured with liquid chromatography coupled with tandem mass spectrometry. A continuous rat intestine absorption model was extended to include drug dissolution and was used to predict the absorption kinetics of GLY depending on particle size. Additional literature datasets of rat GLY formulations with particle sizes ranging from 0.25 to 4.0 μm were used for model predictions. The model predicted reasonably well the absorption profiles of GLY based on varying particle size and varying feeding time. The model predicted inhibition of intestinal uptake or efflux transporters depending on the datasets. The three datasets used formulations with different excipients, which may impact the transporter activity. Model simulations indicated that the model provides a facile framework to predict the impact of transporter inhibition on drug C-t profiles. Model simulations can also be conducted to evaluate the impact of an altered intestinal lumen environment. In conclusion, the rat continuous intestine absorption model may provide a useful tool to predict the impact of varying drug formulations on rat oral absorption profiles.
Collapse
Affiliation(s)
- Swati Nagar
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania19140, United States
| | - Casey Radice
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania19140, United States
| | - Robert Tuohy
- Pace Analytical Life Sciences LLC, Norristown, Pennsylvania19401, United States
| | - Raymond Stevens
- Particle Solutions LLC, West Chester, Pennsylvania19382, United States
| | - Dale Bennyhoff
- Particle Solutions LLC, West Chester, Pennsylvania19382, United States
| | - Ken Korzekwa
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, Pennsylvania19140, United States
| |
Collapse
|
3
|
Garner BR, Stolarz AJ, Stuckey D, Sarimollaoglu M, Liu Y, Palade PT, Rusch NJ, Mu S. K ATP Channel Openers Inhibit Lymphatic Contractions and Lymph Flow as a Possible Mechanism of Peripheral Edema. J Pharmacol Exp Ther 2021; 376:40-50. [PMID: 33100270 PMCID: PMC7745085 DOI: 10.1124/jpet.120.000121] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/08/2020] [Indexed: 11/22/2022] Open
Abstract
Pharmacological openers of ATP-sensitive potassium (KATP) channels are effective antihypertensive agents, but off-target effects, including severe peripheral edema, limit their clinical usefulness. It is presumed that the arterial dilation induced by KATP channel openers (KCOs) increases capillary pressure to promote filtration edema. However, KATP channels also are expressed by lymphatic muscle cells (LMCs), raising the possibility that KCOs also attenuate lymph flow to increase interstitial fluid. The present study explored the effect of KCOs on lymphatic contractile function and lymph flow. In isolated rat mesenteric lymph vessels (LVs), the prototypic KATP channel opener cromakalim (0.01-3 µmol/l) progressively inhibited rhythmic contractions and calculated intraluminal flow. Minoxidil sulfate and diazoxide (0.01-100 µmol/l) had similar effects at clinically relevant plasma concentrations. High-speed in vivo imaging of the rat mesenteric lymphatic circulation revealed that superfusion of LVs with cromakalim and minoxidil sulfate (0.01-10 µmol/l) maximally decreased lymph flow in vivo by 38.4% and 27.4%, respectively. Real-time polymerase chain reaction and flow cytometry identified the abundant KATP channel subunits in LMCs as the pore-forming Kir6.1/6.2 and regulatory sulfonylurea receptor 2 subunits. Patch-clamp studies detected cromakalim-elicited unitary K+ currents in cell-attached patches of LMCs with a single-channel conductance of 46.4 pS, which is a property consistent with Kir6.1/6.2 tetrameric channels. Addition of minoxidil sulfate and diazoxide elicited unitary currents of similar amplitude. Collectively, our findings indicate that KCOs attenuate lymph flow at clinically relevant plasma concentrations as a potential contributing mechanism to peripheral edema. SIGNIFICANCE STATEMENT: ATP-sensitive potassium (KATP) channel openers (KCOs) are potent antihypertensive medications, but off-target effects, including severe peripheral edema, limit their clinical use. Here, we demonstrate that KCOs impair the rhythmic contractions of lymph vessels and attenuate lymph flow, which may promote edema formation. Our finding that the KATP channels in lymphatic muscle cells may be unique from their counterparts in arterial muscle implies that designing arterial-selective KCOs may avoid activation of lymphatic KATP channels and peripheral edema.
Collapse
Affiliation(s)
- Brittney R Garner
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Amanda J Stolarz
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Daniel Stuckey
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Mustafa Sarimollaoglu
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Yunmeng Liu
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Philip T Palade
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Nancy J Rusch
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Shengyu Mu
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
4
|
Cui G, Hong J, Chung-Davidson YW, Infield D, Xu X, Li J, Simhaev L, Khazanov N, Stauffer B, Imhoff B, Cottrill K, Blalock JE, Li W, Senderowitz H, Sorscher E, McCarty NA, Gaggar A. An Ancient CFTR Ortholog Informs Molecular Evolution in ABC Transporters. Dev Cell 2019; 51:421-430.e3. [PMID: 31679858 DOI: 10.1016/j.devcel.2019.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 07/30/2019] [Accepted: 09/24/2019] [Indexed: 01/13/2023]
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is a chloride channel central to the development of secretory diarrhea and cystic fibrosis. The oldest CFTR ortholog identified is from dogfish shark, which retains similar structural and functional characteristics to the mammalian protein, thereby highlighting CFTR's critical role in regulating epithelial ion transport in vertebrates. However, the identification of an early CFTR ortholog with altered structure or function would provide critical insight into the evolution of epithelial anion transport. Here, we describe the earliest known CFTR, expressed in sea lamprey (Petromyzon marinus), with unique structural features, altered kinetics of activation and sensitivity to inhibition, and altered single-channel conductance compared to human CFTR. Our data provide the earliest evolutionary evidence of CFTR, offering insight regarding changes in gene and protein structure that underpin evolution from transporter to anion channel. Importantly, these data provide a unique platform to enhance our understanding of vertebrate phylogeny over a critical period of evolutionary expansion.
Collapse
Affiliation(s)
- Guiying Cui
- Department of Pediatrics and Children's Healthcare of Atlanta, Center for Cystic Fibrosis and Airways Disease Research, Emory University, Atlanta, GA 30322, USA
| | - Jeong Hong
- Department of Pediatrics and Children's Healthcare of Atlanta, Center for Cystic Fibrosis and Airways Disease Research, Emory University, Atlanta, GA 30322, USA; Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, and Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yu-Wen Chung-Davidson
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI 48823, USA
| | - Daniel Infield
- Department of Pediatrics and Children's Healthcare of Atlanta, Center for Cystic Fibrosis and Airways Disease Research, Emory University, Atlanta, GA 30322, USA; Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA
| | - Xin Xu
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, and Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Birmingham Veterans Administration Medical Center, Birmingham, AL 35233, USA
| | - Jindong Li
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, and Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Birmingham Veterans Administration Medical Center, Birmingham, AL 35233, USA
| | - Luba Simhaev
- Department of Chemistry, Bar-Ilan University, Ramat Gan, Israel
| | - Netaly Khazanov
- Department of Chemistry, Bar-Ilan University, Ramat Gan, Israel
| | - Brandon Stauffer
- Department of Pediatrics and Children's Healthcare of Atlanta, Center for Cystic Fibrosis and Airways Disease Research, Emory University, Atlanta, GA 30322, USA
| | - Barry Imhoff
- Department of Pediatrics and Children's Healthcare of Atlanta, Center for Cystic Fibrosis and Airways Disease Research, Emory University, Atlanta, GA 30322, USA
| | - Kirsten Cottrill
- Department of Pediatrics and Children's Healthcare of Atlanta, Center for Cystic Fibrosis and Airways Disease Research, Emory University, Atlanta, GA 30322, USA
| | - J Edwin Blalock
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, and Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Weiming Li
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI 48823, USA
| | | | - Eric Sorscher
- Department of Pediatrics and Children's Healthcare of Atlanta, Center for Cystic Fibrosis and Airways Disease Research, Emory University, Atlanta, GA 30322, USA; Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, and Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Nael A McCarty
- Department of Pediatrics and Children's Healthcare of Atlanta, Center for Cystic Fibrosis and Airways Disease Research, Emory University, Atlanta, GA 30322, USA
| | - Amit Gaggar
- Department of Medicine, Gregory Fleming James Cystic Fibrosis Research Center, and Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Birmingham Veterans Administration Medical Center, Birmingham, AL 35233, USA.
| |
Collapse
|
5
|
Martin GM, Sung MW, Yang Z, Innes LM, Kandasamy B, David LL, Yoshioka C, Shyng SL. Mechanism of pharmacochaperoning in a mammalian K ATP channel revealed by cryo-EM. eLife 2019; 8:46417. [PMID: 31343405 PMCID: PMC6699824 DOI: 10.7554/elife.46417] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/22/2019] [Indexed: 01/03/2023] Open
Abstract
ATP-sensitive potassium (KATP) channels composed of a pore-forming Kir6.2 potassium channel and a regulatory ABC transporter sulfonylurea receptor 1 (SUR1) regulate insulin secretion in pancreatic β-cells to maintain glucose homeostasis. Mutations that impair channel folding or assembly prevent cell surface expression and cause congenital hyperinsulinism. Structurally diverse KATP inhibitors are known to act as pharmacochaperones to correct mutant channel expression, but the mechanism is unknown. Here, we compare cryoEM structures of a mammalian KATP channel bound to pharmacochaperones glibenclamide, repaglinide, and carbamazepine. We found all three drugs bind within a common pocket in SUR1. Further, we found the N-terminus of Kir6.2 inserted within the central cavity of the SUR1 ABC core, adjacent the drug binding pocket. The findings reveal a common mechanism by which diverse compounds stabilize the Kir6.2 N-terminus within SUR1’s ABC core, allowing it to act as a firm ‘handle’ for the assembly of metastable mutant SUR1-Kir6.2 complexes.
Collapse
Affiliation(s)
- Gregory M Martin
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, United States
| | - Min Woo Sung
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, United States
| | - Zhongying Yang
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, United States
| | - Laura M Innes
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, United States
| | - Balamurugan Kandasamy
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, United States
| | - Larry L David
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, United States
| | - Craig Yoshioka
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, United States
| | - Show-Ling Shyng
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, United States
| |
Collapse
|
6
|
David M, Lebrun C, Duguet T, Talmont F, Beech R, Orlowski S, André F, Prichard RK, Lespine A. Structural model, functional modulation by ivermectin and tissue localization of Haemonchus contortus P-glycoprotein-13. Int J Parasitol Drugs Drug Resist 2018; 8:145-157. [PMID: 29571165 PMCID: PMC6114108 DOI: 10.1016/j.ijpddr.2018.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/09/2018] [Accepted: 02/12/2018] [Indexed: 12/18/2022]
Abstract
Haemonchus contortus, one of the most economically important parasites of small ruminants, has become resistant to the anthelmintic ivermectin. Deciphering the role of P-glycoproteins in ivermectin resistance is desirable for understanding and overcoming this resistance. In the model nematode, Caenorhabditis elegans, P-glycoprotein-13 is expressed in the amphids, important neuronal structures for ivermectin activity. We have focused on its ortholog in the parasite, Hco-Pgp-13. A 3D model of Hco-Pgp-13, presenting an open inward-facing conformation, has been constructed by homology with the Cel-Pgp-1 crystal structure. In silico docking calculations predicted high affinity binding of ivermectin and actinomycin D to the inner chamber of the protein. Following in vitro expression, we showed that ivermectin and actinomycin D modulated Hco-Pgp-13 ATPase activity with high affinity. Finally, we found in vivo Hco-Pgp-13 localization in epithelial, pharyngeal and neuronal tissues. Taken together, these data suggest a role for Hco-Pgp-13 in ivermectin transport, which could contribute to anthelmintic resistance.
Collapse
Affiliation(s)
- Marion David
- INTHERES, Université de Toulouse, INRA, ENVT, Toulouse, France; Institute of Parasitology, McGill University, Sainte-Anne-De-Bellevue, Canada
| | - Chantal Lebrun
- INTHERES, Université de Toulouse, INRA, ENVT, Toulouse, France
| | - Thomas Duguet
- Institute of Parasitology, McGill University, Sainte-Anne-De-Bellevue, Canada
| | - Franck Talmont
- Institute of Pharmacology and Structural Biology, UMR 5089, CNRS, Toulouse, France
| | - Robin Beech
- Institute of Parasitology, McGill University, Sainte-Anne-De-Bellevue, Canada
| | - Stéphane Orlowski
- CEA, Institut de Biologie Frédéric Joliot, Centre de Saclay, SB2SM, UMR9198 CNRS, I2BC; 91191 Gif-sur-Yvette Cedex, France
| | - François André
- CEA, Institut de Biologie Frédéric Joliot, Centre de Saclay, SB2SM, UMR9198 CNRS, I2BC; 91191 Gif-sur-Yvette Cedex, France
| | - Roger K Prichard
- Institute of Parasitology, McGill University, Sainte-Anne-De-Bellevue, Canada.
| | - Anne Lespine
- INTHERES, Université de Toulouse, INRA, ENVT, Toulouse, France.
| |
Collapse
|
7
|
Population pharmacokinetics of carvedilol enantiomers and their metabolites in healthy subjects and type-2 diabetes patients. Eur J Pharm Sci 2017; 109S:S108-S115. [DOI: 10.1016/j.ejps.2017.05.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 05/15/2017] [Indexed: 12/20/2022]
|
8
|
Zhu XC, Zhou K, Xu SQ, Ma YB. Diagnostic Value of Semiquantitative Analysis of 99mTechnetium-Methoxyisobutylisonitrile (99mTc-MIBI) Imaging in Predicting Early-Stage Cervical Lymph Node Metastasis of Thyroid Carcinoma. Med Sci Monit 2017; 23:1552-1558. [PMID: 28362720 PMCID: PMC5386442 DOI: 10.12659/msm.899966] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background The aim of this study was to evaluate the value of semiquantitative analysis (SQA) of 99mTc-MIBI imaging in predicting early-stage cervical lymph node metastasis (CLNM) in thyroid carcinoma (TC). Material/Methods TC patients (n=106) undergoing surgical resection and histopathological examination were enrolled. All patients received 99mTc-MIBI imaging prior to surgery. P-glycoprotein (P-gp) expression was detected by PT-PCR and immunohistochemistry. With pathological results as the criterion standard, the diagnostic efficiency of 99mTc-MIBI imaging in predicting early-stage CLNM was evaluated. The correlation of P-gp with 99mTc-MIBI imaging was investigated. Logistic regression analysis was applied for analyzing the factors affecting early-stage CLNM. Results The detection rate and misdiagnosis rate of 99mTc-MIBI imaging for early-stage CLNM diagnosis were 87.3% and 12.7%, respectively. Receiver operating characteristic (ROC) curve analysis showed an accuracy of 99mTc-MIBI imaging of 85.85%. Preoperative 99mTc-MIBI scan showed statistical differences between metastasis and non-metastasis groups in early and delayed T/NT and washout rate (all P<0.05). The percentage of P-gp-expressing cells and the expression rate of P-gp gene both exhibited statistical differences between metastasis and no-metastasis groups (both P<0.05). Tumor diameter, lesion distribution, the percentage of P-gp-expressing cells, and the expression rate of P-gp gene were risk factors for CLNM (all P<0.05). Conclusions 99mTc-MIBI imaging has value in qualitative diagnosis of early-stage CLNM in TC. Tumor diameter, lesion distribution, the percentage of P-gp-expressing cells, and the expression rate of P-gp gene were risk factors for CLNM.
Collapse
Affiliation(s)
- Xiao-Chun Zhu
- Department of Nuclear Medicine, Shanghai 9th People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (mainland)
| | - Kai Zhou
- Department of Laboratory, Shanghai 9th People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (mainland)
| | - Shi-Qing Xu
- Department of Nuclear Medicine, Shanghai 9th People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (mainland)
| | - Yu-Bo Ma
- Department of Nuclear Medicine, Shanghai 9th People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China (mainland)
| |
Collapse
|
9
|
Nardotto GHB, Coelho EB, Paiva CE, Lanchote VL. Effects of Type 2 Diabetes Mellitus in Patients on Treatment With Glibenclamide and Metformin on Carvedilol Enantiomers Metabolism. J Clin Pharmacol 2017; 57:760-769. [DOI: 10.1002/jcph.864] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 11/30/2016] [Indexed: 11/09/2022]
Affiliation(s)
- Glauco H. B. Nardotto
- Faculdade de Ciências Farmacêuticas Ribeirão Preto; Universidade de São Paulo; São Paulo Brazil
| | - Eduardo B. Coelho
- Faculdade de Medicina de Ribeirão Preto; Universidade de São Paulo; São Paulo Brazil
| | - Carlos E. Paiva
- Faculdade de Medicina de Ribeirão Preto; Universidade de São Paulo; São Paulo Brazil
| | - Vera L. Lanchote
- Faculdade de Ciências Farmacêuticas Ribeirão Preto; Universidade de São Paulo; São Paulo Brazil
| |
Collapse
|
10
|
László L, Sarkadi B, Hegedűs T. Jump into a New Fold-A Homology Based Model for the ABCG2/BCRP Multidrug Transporter. PLoS One 2016; 11:e0164426. [PMID: 27741279 PMCID: PMC5065228 DOI: 10.1371/journal.pone.0164426] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 09/23/2016] [Indexed: 12/03/2022] Open
Abstract
ABCG2/BCRP is a membrane protein, involved in xenobiotic and endobiotic transport in key pharmacological barriers and drug metabolizing organs, in the protection of stem cells, and in multidrug resistance of cancer. Pharmacogenetic studies implicated the role of ABCG2 in response to widely used medicines and anticancer agents, as well as in gout. Its Q141K variant exhibits decreased functional expression thus increased drug accumulation and decreased urate secretion. Still, there has been no reliable molecular model available for this protein, as the published structures of other ABC transporters could not be properly fitted to the ABCG2 topology and experimental data. The recently published high resolution structure of a close homologue, the ABCG5-ABCG8 heterodimer, revealed a new ABC transporter fold, unique for ABCG proteins. Here we present a structural model of the ABCG2 homodimer based on this fold and detail the experimental results supporting this model. In order to describe the effect of mutations on structure and dynamics, and characterize substrate recognition and cholesterol regulation we performed molecular dynamics simulations using full length ABCG2 protein embedded in a membrane bilayer and in silico docking simulations. Our results show that in the Q141K variant the introduced positive charge diminishes the interaction between the nucleotide binding and transmembrane domains and the R482G variation alters the orientation of transmembrane helices. Moreover, the R482 position, which plays a role the substrate specificity of the transporter, is located in one of the substrate binding pockets identified by the in silico docking calculations. In summary, the ABCG2 model and in silico simulations presented here may have significant impact on understanding drug distribution and toxicity, as well as drug development against cancer chemotherapy resistance or gout.
Collapse
Affiliation(s)
- Laura László
- Molecular Biophysics Research Group and Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Balázs Sarkadi
- Molecular Biophysics Research Group and Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
- Research Centre for Natural Sciences, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary
| | - Tamás Hegedűs
- Molecular Biophysics Research Group and Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
11
|
In silico analysis of the binding of anthelmintics to Caenorhabditis elegansP-glycoprotein 1. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2016; 6:299-313. [PMID: 27746191 PMCID: PMC5196494 DOI: 10.1016/j.ijpddr.2016.09.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 09/07/2016] [Accepted: 09/09/2016] [Indexed: 11/23/2022]
Abstract
Macrocyclic lactones (ML) are important anthelmintics used in animals and humans against parasite nematodes, but their therapeutic success is compromised by the spread of ML resistance. Some ABC transporters, such as p-glycoproteins (Pgps), are selected and overexpressed in ML-resistant nematodes, supporting a role for some drug efflux proteins in ML resistance. However, the role of such proteins in ML transport remains to be clarified at the molecular level. Recently, Caenorhabditis elegans Pgp-1 (Cel-Pgp-1) has been crystallized, and its drug-modulated ATPase function characterized in vitro revealed Cel-Pgp-1 as a multidrug transporter. Using this crystal structure, we have developed an in silico drug docking model in order to study the binding of ML and other anthelmintic drugs to Cel-Pgp-1. All tested ML bound with high affinity in a unique site, within the inner chamber of the protein, supporting that ML may be transported by Cel-Pgp-1. Interestingly, interacting residues delineate a ML specific fingerprint involving H-bonds, including T1028. In particular, benzofurane and spiroketal moieties bound to specific sub-sites. When compared with the aglycone ML, such as moxidectin and ivermectin aglycone, avermectin anthelmintics have significant higher affinity for Cel-Pgp-1, likely due to the sugar substituent(s) that bind to a specific area involving H-bonds at Y771. Triclabendazole, closantel and emodepside bound with good affinities to different sub-sites in the inner chamber, partially overlapping with the ML binding site, suggesting that they could compete for Cel-Pgp-1-mediated ML transport. In conclusion, this work provides novel information on the role of nematode Pgps in transporting anthelmintics, and a valuable tool to predict drug-drug interactions and to rationally design new competitive inhibitors of clinically-relevant nematode Pgps, to improve anthelmintic therapeutics.
Collapse
|
12
|
The shifting landscape of KATP channelopathies and the need for 'sharper' therapeutics. Future Med Chem 2016; 8:789-802. [PMID: 27161588 DOI: 10.4155/fmc-2016-0005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
ATP-sensitive potassium (KATP) channels play fundamental roles in the regulation of endocrine, neural and cardiovascular function. Small-molecule inhibitors (e.g., sulfonylurea drugs) or activators (e.g., diazoxide) acting on SUR1 or SUR2 have been used clinically for decades to manage the inappropriate secretion of insulin in patients with Type 2 diabetes, hyperinsulinism and intractable hypertension. More recently, the discovery of rare disease-causing mutations in KATP channel-encoding genes has highlighted the need for new therapeutics for the treatment of certain forms of neonatal diabetes mellitus, congenital hyperinsulinism and Cantu syndrome. Here, we provide a high-level overview of the pathophysiology of these diseases and discuss the development of a flexible high-throughput screening platform to enable the development of new classes of KATP channel modulators.
Collapse
|
13
|
Principalli MA, Dupuis JP, Moreau CJ, Vivaudou M, Revilloud J. Kir6.2 activation by sulfonylurea receptors: a different mechanism of action for SUR1 and SUR2A subunits via the same residues. Physiol Rep 2015; 3:3/9/e12533. [PMID: 26416970 PMCID: PMC4600379 DOI: 10.14814/phy2.12533] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
ATP-sensitive potassium channels (K-ATP channels) play a key role in adjusting the membrane potential to the metabolic state of cells. They result from the unique combination of two proteins: the sulfonylurea receptor (SUR), an ATP-binding cassette (ABC) protein, and the inward rectifier K(+) channel Kir6.2. Both subunits associate to form a heterooctamer (4 SUR/4 Kir6.2). SUR modulates channel gating in response to the binding of nucleotides or drugs and Kir6.2 conducts potassium ions. The activity of K-ATP channels varies with their localization. In pancreatic β-cells, SUR1/Kir6.2 channels are partly active at rest while in cardiomyocytes SUR2A/Kir6.2 channels are mostly closed. This divergence of function could be related to differences in the interaction of SUR1 and SUR2A with Kir6.2. Three residues (E1305, I1310, L1313) located in the linker region between transmembrane domain 2 and nucleotide-binding domain 2 of SUR2A were previously found to be involved in the activation pathway linking binding of openers onto SUR2A and channel opening. To determine the role of the equivalent residues in the SUR1 isoform, we designed chimeras between SUR1 and the ABC transporter multidrug resistance-associated protein 1 (MRP1), and used patch clamp recordings on Xenopus oocytes to assess the functionality of SUR1/MRP1 chimeric K-ATP channels. Our results reveal that the same residues in SUR1 and SUR2A are involved in the functional association with Kir6.2, but they display unexpected side-chain specificities which could account for the contrasted properties of pancreatic and cardiac K-ATP channels.
Collapse
Affiliation(s)
- Maria A Principalli
- Institut de Biologie Structurale (IBS), University of Grenoble Alpes, Grenoble, France CNRS, IBS, Grenoble, France CEA, IBS, Grenoble, France
| | - Julien P Dupuis
- Institut Interdisciplinaire de Neurosciences CNRS UMR 5297, Bordeaux, France
| | - Christophe J Moreau
- Institut de Biologie Structurale (IBS), University of Grenoble Alpes, Grenoble, France CNRS, IBS, Grenoble, France CEA, IBS, Grenoble, France
| | - Michel Vivaudou
- Institut de Biologie Structurale (IBS), University of Grenoble Alpes, Grenoble, France CNRS, IBS, Grenoble, France CEA, IBS, Grenoble, France
| | - Jean Revilloud
- Institut de Biologie Structurale (IBS), University of Grenoble Alpes, Grenoble, France CNRS, IBS, Grenoble, France CEA, IBS, Grenoble, France
| |
Collapse
|
14
|
Systemic Administration of Glibenclamide Fails to Achieve Therapeutic Levels in the Brain and Cerebrospinal Fluid of Rodents. PLoS One 2015. [PMID: 26225433 PMCID: PMC4520580 DOI: 10.1371/journal.pone.0134476] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Activating mutations in the Kir6.2 (KCNJ11) subunit of the ATP-sensitive potassium channel cause neonatal diabetes (ND). Patients with severe mutations also suffer from neurological complications. Glibenclamide blocks the open KATP channels and is the treatment of choice for ND. However, although glibenclamide successfully restores normoglycaemia, it has a far more limited effect on the neurological problems. To assess the extent to which glibenclamide crosses the blood-brain barrier (BBB) in vivo, we quantified glibenclamide concentrations in plasma, cerebrospinal fluid (CSF), and brain tissue of rats, control mice, and mice expressing a human neonatal diabetes mutation (Kir6.2-V59M) selectively in neurones (nV59M mice). As only small sample volumes can be obtained from rodents, we developed a highly sensitive method of analysis, using liquid chromatography tandem mass spectrometry acquisition with pseudo-selected reaction monitoring, achieving a quantification limit of 10ng/ml (20nM) glibenclamide in a 30μl sample. Glibenclamide was not detectable in the CSF or brain of rats after implantation with subcutaneous glibenclamide pellets, despite high plasma concentrations. Further, one hour after a suprapharmacological glibenclamide dose was administered directly into the lateral ventricle of the brain, the plasma concentration was twice that of the CSF. This suggests the drug is rapidly exported from the CSF. Elacridar, an inhibitor of P-glycoprotein and breast cancer resistance protein (major multidrug resistance transporters at the BBB), did not affect glibenclamide levels in CSF and brain tissue. We also identified a reduced sensitivity to volatile anaesthetics in nV59M mice and showed this was not reversed by systemic delivery of glibenclamide. Our results therefore suggest that little glibenclamide reaches the central nervous system when given systemically, that glibenclamide is rapidly removed across the BBB when given intracranioventricularly, and that any glibenclamide that does enter (and is below our detection limit) is insufficient to influence neuronal function as assessed by anaesthesia sensitivity.
Collapse
|
15
|
Brayer SW, Reddy KR. Ritonavir-boosted protease inhibitor based therapy: a new strategy in chronic hepatitis C therapy. Expert Rev Gastroenterol Hepatol 2015; 9:547-58. [PMID: 25846301 DOI: 10.1586/17474124.2015.1032938] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chronic hepatitis C virus (HCV) infection is a worldwide health issue. All oral therapies are quickly replacing peg-interferon-based treatment regimens. Developing effective, well tolerated, treatments accessible for difficult to treat populations remains an unmet need. Ritonavir, an HIV-1 protease inhibitor, has pharmacokinetic properties that enhance the activity of concomitantly administered direct acting antivirals against HCV. Ritonavir inhibits Cytochrome P450 isozyme 3A4, diminishing first pass effect and hepatic metabolism, changing the pharmacokinetic parameters of Cytochrome P450 isozyme 3A4 substrates. When combined with the HCV protease inhibitor paritaprevir, ritonavir increases mean area under the curve, allowing once daily dosing. While Phase II and III clinical trials with ritonavir-boosted paritaprevir, ombitasvir, and dasabuvir demonstrated high efficacy in those with HCV infection, drug-drug interactions warrant cautious use of ritonavir in specific patient populations. Consideration of the patients' full medication list is imperative due to the ubiquitous nature of the Cytochrome P450 isozyme 3A4 system.
Collapse
Affiliation(s)
- Samuel W Brayer
- University of Pennsylvania, 2 Dulles, 3400 Spruce Street, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
16
|
Devaraneni PK, Martin GM, Olson EM, Zhou Q, Shyng SL. Structurally distinct ligands rescue biogenesis defects of the KATP channel complex via a converging mechanism. J Biol Chem 2015; 290:7980-91. [PMID: 25637631 DOI: 10.1074/jbc.m114.634576] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Small molecules that correct protein misfolding and misprocessing defects offer a potential therapy for numerous human diseases. However, mechanisms underlying pharmacological correction of such defects, especially in heteromeric complexes with structurally diverse constituent proteins, are not well understood. Here we investigate how two chemically distinct compounds, glibenclamide and carbamazepine, correct biogenesis defects in ATP-sensitive potassium (KATP) channels composed of sulfonylurea receptor 1 (SUR1) and Kir6.2. We present evidence that despite structural differences, carbamazepine and glibenclamide compete for binding to KATP channels, and both drugs share a binding pocket in SUR1 to exert their effects. Moreover, both compounds engage Kir6.2, in particular the distal N terminus of Kir6.2, which is involved in normal channel biogenesis, for their chaperoning effects on SUR1 mutants. Conversely, both drugs can correct channel biogenesis defects caused by Kir6.2 mutations in a SUR1-dependent manner. Using an unnatural, photocross-linkable amino acid, azidophenylalanine, genetically encoded in Kir6.2, we demonstrate in living cells that both drugs promote interactions between the distal N terminus of Kir6.2 and SUR1. These findings reveal a converging pharmacological chaperoning mechanism wherein glibenclamide and carbamazepine stabilize the heteromeric subunit interface critical for channel biogenesis to overcome defective biogenesis caused by mutations in individual subunits.
Collapse
Affiliation(s)
- Prasanna K Devaraneni
- From the Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon 97239
| | - Gregory M Martin
- From the Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon 97239
| | - Erik M Olson
- From the Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon 97239
| | - Qing Zhou
- From the Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon 97239
| | - Show-Ling Shyng
- From the Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, Oregon 97239
| |
Collapse
|
17
|
Mahendran G, Manoj M, Murugesh E, Sathish Kumar R, Shanmughavel P, Rajendra Prasad KJ, Narmatha Bai V. In vivo anti-diabetic, antioxidant and molecular docking studies of 1, 2, 8-trihydroxy-6-methoxy xanthone and 1, 2-dihydroxy-6-methoxyxanthone-8-O-β-D-xylopyranosyl isolated from Swertia corymbosa. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2014; 21:1237-1248. [PMID: 25172785 DOI: 10.1016/j.phymed.2014.06.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 04/30/2014] [Accepted: 06/19/2014] [Indexed: 06/03/2023]
Abstract
1, 2, 8-trihydroxy-6-methoxy xanthone (1) and 1, 2- dihydroxy-6-methoxyxanthone-8-O-β-d-xylopyranosyl (2) are the main constituents of petroleum ether and ethyl acetate extracts from Swertia corymbosa (Gentinaceae), a medicinal plant used in Indian traditional system for the treatment of diabetes. The present study was designed to examine the antihypoglycemic, antihyperlipidemic and antioxidant effect of compounds 1 and 2 in streptozotocin (STZ) induced diabetic rats. Diabetes was induced in male Wistar rats by a single intraperitoneal injection of STZ (60 mg/kg b.w.). The isolated compounds 1 and 2 at a dose of 50 mg/kg b.w., produced the maximum fall of 83% in the blood glucose level in the diabetic rats after 3h of the treatment. The administration of 1 and 2 (50 mg/kgb.w.) daily for 28 days in STZ induced diabetic rats, resulted in a significant decrease in blood glucose, glycosylated hemoglobin, SGOT, SGPT, ALP serum urea and creatinine with significant rise in plasma insulin level. Test compounds 1 and 2 showed antihyperlipidemic activities as evidenced by significant decrease in serum TC, TG, LDL-C, VLDL-C levels coupled together with elevation of HDL-C level in diabetic treated rats when compared to diabetic untreated rats, indicate the protective role against liver and kidney damage. The results of histopathology also showed 1 and 2 protected tissues (pancreas, liver and kidney) against peroxidation damage and maintained tissue integrity. Further, the molecular interaction study of the ligands 1, 2 and glibenclamide with various diabetes mellitus related protein targets like glucokinase (PDB ID: 1V4S), fructose-1, 6-bisphosphatase 1 (PDB ID: 2JJK) 11-β-hydroxysteroid dehydrogenase (PDB ID: 2BEL) and modeled protein sulfonylurea receptor 1 (SUR1) showed that ligand 1 and 2 possess binding affinity with all protein targets except for 2BEL target protein for which ligand 1 has no interaction. The ligand pose with 2BEL and SUR1 protein target of ligand 2 gave the best binding conformation. Hence 1 and 2 can be considered for developing into a potent antidiabetic drug.
Collapse
Affiliation(s)
- G Mahendran
- Plant Tissue Culture Laboratory, Department of Botany, School of Life Sciences, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India.
| | - M Manoj
- Department of Chemistry, School of Chemical Sciences, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - E Murugesh
- Computational Biology Lab, Bioinformatics Department, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - R Sathish Kumar
- Computational Biology Lab, Bioinformatics Department, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - P Shanmughavel
- Computational Biology Lab, Bioinformatics Department, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - K J Rajendra Prasad
- Department of Chemistry, School of Chemical Sciences, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - V Narmatha Bai
- Plant Tissue Culture Laboratory, Department of Botany, School of Life Sciences, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| |
Collapse
|
18
|
Hosy E, Vivaudou M. The unusual stoichiometry of ADP activation of the KATP channel. Front Physiol 2014; 5:11. [PMID: 24478723 PMCID: PMC3904077 DOI: 10.3389/fphys.2014.00011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/07/2014] [Indexed: 11/27/2022] Open
Abstract
KATP channels, oligomers of 4 pore-forming Kir6.2 proteins and 4 sulfonylurea receptors (SUR), sense metabolism by monitoring both cytosolic ATP, which closes the channel by interacting with Kir6.2, and ADP, which opens it via SUR. SUR mutations that alter activation by ADP are a major cause of KATP channelopathies. We examined the mechanism of ADP activation by analysis of single-channel and macropatch recordings from Xenopus oocytes expressing various mixtures of wild-type SUR2A and an ADP-activation-defective mutant. Evaluation of the data by a binomial distribution model suggests that wild-type and mutant SURs freely co-assemble and that channel activation results from interaction of ADP with only 2 of 4 SURs. This finding explains the heterozygous nature of most KATP channelopathies linked to mutations altering ADP activation. It also suggests that the channel deviates from circular symmetry and could function as a dimer-of-dimers.
Collapse
Affiliation(s)
- Eric Hosy
- Institut de Biologie Structurale, University Grenoble Alpes Grenoble, France ; Laboratory of Excellence, Ion Channel Science and Therapeutics, CNRS, Institut de Biologie Structurale Grenoble, France ; CEA, DSV, Institut de Biologie Structurale Grenoble, France
| | - Michel Vivaudou
- Institut de Biologie Structurale, University Grenoble Alpes Grenoble, France ; Laboratory of Excellence, Ion Channel Science and Therapeutics, CNRS, Institut de Biologie Structurale Grenoble, France ; CEA, DSV, Institut de Biologie Structurale Grenoble, France
| |
Collapse
|
19
|
It Takes More Than Two to Tango: Regulation of Plant ABC Transporters. SIGNALING AND COMMUNICATION IN PLANTS 2014. [DOI: 10.1007/978-3-319-06511-3_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
20
|
Wen PC, Verhalen B, Wilkens S, Mchaourab HS, Tajkhorshid E. On the origin of large flexibility of P-glycoprotein in the inward-facing state. J Biol Chem 2013; 288:19211-20. [PMID: 23658020 PMCID: PMC3696692 DOI: 10.1074/jbc.m113.450114] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
P-glycoprotein (Pgp) is one of the most biomedically relevant transporters in the ATP binding
cassette (ABC) superfamily due to its involvement in developing multidrug resistance in cancer
cells. Employing molecular dynamics simulations and double electron-electron resonance spectroscopy,
we have investigated the structural dynamics of membrane-bound Pgp in the inward-facing state and
found that Pgp adopts an unexpectedly wide range of conformations, highlighted by the degree of
separation between the two nucleotide-binding domains (NBDs). The distance between the two NBDs in
the equilibrium simulations covers a range of at least 20 Å, including, both, more open and
more closed NBD configurations than the crystal structure. The double electron-electron resonance
measurements on spin-labeled Pgp mutants also show wide distributions covering both longer and
shorter distances than those observed in the crystal structure. Based on structural and sequence
analyses, we propose that the transmembrane domains of Pgp might be more flexible than other
structurally known ABC exporters. The structural flexibility of Pgp demonstrated here is not only in
close agreement with, but also helps rationalize, the reported high NBD fluctuations in several ABC
exporters and possibly represents a fundamental difference in the transport mechanism between ABC
exporters and ABC importers. In addition, during the simulations we have captured partial entrance
of a lipid molecule from the bilayer into the lumen of Pgp, reaching the putative drug binding site.
The location of the protruding lipid suggests a putative pathway for direct drug recruitment from
the membrane.
Collapse
Affiliation(s)
- Po-Chao Wen
- Center for Biophysics and Computational Biology, Department of Biochemistry, College of Medicine, and The Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illnois 61801, USA
| | | | | | | | | |
Collapse
|
21
|
Simard JM, Woo SK, Aarabi B, Gerzanich V. The Sur1-Trpm4 Channel in Spinal Cord Injury. ACTA ACUST UNITED AC 2013; Suppl 4. [PMID: 24834370 DOI: 10.4172/2165-7939.s4-002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Spinal cord injury (SCI) is a major unsolved challenge in medicine. Impact trauma to the spinal cord shears blood vessels, causing an immediate 'primary hemorrhage'. During the hours following trauma, the region of hemorrhage enlarges progressively, with delayed or 'secondary hemorrhage' adding to the primary hemorrhage, and effectively doubling its volume. The process responsible for the secondary hemorrhage that results in early expansion of the hemorrhagic lesion is termed 'progressive hemorrhagic necrosis' (PHN). PHN is a dynamic process of auto destruction whose molecular underpinnings are only now beginning to be elucidated. PHN results from the delayed, progressive, catastrophic failure of the structural integrity of capillaries. The resulting 'capillary fragmentation' is a unique, pathognomonic feature of PHN. Recent work has implicated the Sur1-Trpm4 channel that is newly upregulated in penumbral microvessels as being required for the development of PHN. Targeting the Sur1-Trpm4 channel by gene deletion, gene suppression, or pharmacological inhibition of either of the two channel subunits, Sur1 or Trpm4, yields exactly the same effects histologically and functionally, and exactly the same unique, pathognomonic phenotype - the prevention of capillary fragmentation. The potential advantage of inhibiting Sur1-Trpm4 channels using glibenclamide is a highly promising strategy for ameliorating the devastating sequelae of spinal cord trauma in humans.
Collapse
Affiliation(s)
- J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, USA ; Department of Pathology, University of Maryland School of Medicine, Baltimore, USA ; Department of Physiology, University of Maryland School of Medicine, Baltimore, USA
| | - Seung Kyoon Woo
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, USA
| | - Bizhan Aarabi
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, USA
| |
Collapse
|
22
|
Corradi V, Singh G, Tieleman DP. The human transporter associated with antigen processing: molecular models to describe peptide binding competent states. J Biol Chem 2012; 287:28099-111. [PMID: 22700967 PMCID: PMC3431710 DOI: 10.1074/jbc.m112.381251] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The human transporter associated with antigen processing (TAP) is a member of the ATP binding cassette (ABC) transporter superfamily. TAP plays an essential role in the antigen presentation pathway by translocating cytosolic peptides derived from proteasomal degradation into the endoplasmic reticulum lumen. Here, the peptides are loaded into major histocompatibility class I molecules to be in turn exposed at the cell surface for recognition by T-cells. TAP is a heterodimer formed by the association of two half-transporters, TAP1 and TAP2, with a typical ABC transporter core that consists of two nucleotide binding domains and two transmembrane domains. Despite the availability of biological data, a full understanding of the mechanism of action of TAP is limited by the absence of experimental structures of the full-length transporter. Here, we present homology models of TAP built on the crystal structures of P-glycoprotein, ABCB10, and Sav1866. The models represent the transporter in inward- and outward-facing conformations that could represent initial and final states of the transport cycle, respectively. We described conserved regions in the endoplasmic reticulum-facing loops with a role in the opening and closing of the cavity. We also identified conserved π-stacking interactions in the cytosolic part of the transmembrane domains that could explain the experimental data available for TAP1-Phe-265. Electrostatic potential calculations gave structural insights into the role of residues involved in peptide binding, such as TAP1-Val-288, TAP2-Cys-213, TAP2-Met-218. Moreover, these calculations identified additional residues potentially involved in peptide binding, in turn verified with replica exchange simulations performed on a peptide bound to the inward-facing models.
Collapse
Affiliation(s)
- Valentina Corradi
- Department of Biological Sciences and Institute for Biocomplexity and Informatics, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | | | | |
Collapse
|
23
|
Simard JM, Tsymbalyuk O, Keledjian K, Ivanov A, Ivanova S, Gerzanich V. Comparative effects of glibenclamide and riluzole in a rat model of severe cervical spinal cord injury. Exp Neurol 2011; 233:566-74. [PMID: 22177998 DOI: 10.1016/j.expneurol.2011.11.044] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 09/21/2011] [Accepted: 11/29/2011] [Indexed: 01/08/2023]
Abstract
Both glibenclamide and riluzole reduce necrosis and improve outcome in rat models of spinal cord injury (SCI). In SCI, gene suppression experiments show that newly upregulated sulfonylurea receptor 1 (Sur1)-regulated NC(Ca-ATP) channels in microvascular endothelial cells are responsible for "persistent sodium currents" that cause capillary fragmentation and "progressive hemorrhagic necrosis". Glibenclamide is a potent blocker of Sur1-regulated NC(Ca-ATP) channels (IC(50), 6-48 nM). Riluzole is a pleotropic drug that blocks "persistent sodium currents" in neurons, but in SCI, its molecular mechanism of action is uncertain. We hypothesized that riluzole might block the putative pore-forming subunits of Sur1-regulated NC(Ca-ATP) channels, Trpm4. In patch clamp experiments, riluzole blocked Sur1-regulated NC(Ca-ATP) channels in endothelial cells and heterologously expressed Trpm4 (IC(50), 31 μM). Using a rat model of cervical SCI associated with high mortality, we compared the effects of glibenclamide and riluzole administered beginning at 3h and continuing for 7 days after impact. During the acute phase, both drugs reduced capillary fragmentation and progressive hemorrhagic necrosis, and both prevented death. At 6 weeks, modified (unilateral) Basso, Beattie, Bresnahan locomotor scores were similar, but measures of complex function (grip strength, rearing, accelerating rotarod) and tissue sparing were significantly better with glibenclamide than with riluzole. We conclude that both drugs act similarly, glibenclamide on the regulatory subunit, and riluzole on the putative pore-forming subunit of the Sur1-regulated NC(Ca-ATP) channel. Differences in specificity, dose-limiting potency, or in spectrum of action may account for the apparent superiority of glibenclamide over riluzole in this model of severe SCI.
Collapse
Affiliation(s)
- J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201-1595, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Stahl M, Stahl K, Brubacher MB, Forrest JN. Divergent CFTR orthologs respond differently to the channel inhibitors CFTRinh-172, glibenclamide, and GlyH-101. Am J Physiol Cell Physiol 2011; 302:C67-76. [PMID: 21940661 DOI: 10.1152/ajpcell.00225.2011] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Comparison of diverse orthologs is a powerful tool to study the structure and function of channel proteins. We investigated the response of human, killifish, pig, and shark cystic fibrosis transmembrane conductance regulator (CFTR) to specific inhibitors of the channel: CFTR(inh)-172, glibenclamide, and GlyH-101. In three systems, including organ perfusion of the shark rectal gland, primary cultures of shark rectal gland tubules, and expression studies of each ortholog in cRNA microinjected Xenopus laevis oocytes, we observed fundamental differences in the sensitivity to inhibition by these channel blockers. In organ perfusion studies, shark CFTR was insensitive to inhibition by CFTR(inh)-172. This insensitivity was also seen in short-circuit current experiments with cultured rectal gland tubular epithelial cells (maximum inhibition 4 ± 1.3%). In oocyte expression studies, shark CFTR was again insensitive to CFTR(inh)-172 (maximum inhibition 10.3 ± 2.5% at 25 μM), pig CFTR was insensitive to glibenclamide (maximum inhibition 18.4 ± 4.4% at 250 μM), and all orthologs were sensitive to GlyH-101. The amino acid residues considered responsible by previous site-directed mutagenesis for binding of the three inhibitors are conserved in the four CFTR isoforms studied. These experiments demonstrate a profound difference in the sensitivity of different orthologs of CFTR proteins to inhibition by CFTR blockers that cannot be explained by mutagenesis of single amino acids. We believe that the potency of the inhibitors CFTR(inh)-172, glibenclamide, and GlyH-101 on the CFTR chloride channel protein is likely dictated by the local environment and the three-dimensional structure of additional residues that form the vestibules, the chloride pore, and regulatory regions of the channel.
Collapse
Affiliation(s)
- Maximilian Stahl
- Nephrology Division, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06510-3222, USA
| | | | | | | |
Collapse
|
25
|
Bailly A, Yang H, Martinoia E, Geisler M, Murphy AS. Plant Lessons: Exploring ABCB Functionality Through Structural Modeling. FRONTIERS IN PLANT SCIENCE 2011; 2:108. [PMID: 22639627 PMCID: PMC3355715 DOI: 10.3389/fpls.2011.00108] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Accepted: 12/17/2011] [Indexed: 05/18/2023]
Abstract
In contrast to mammalian ABCB1 proteins, narrow substrate specificity has been extensively documented for plant orthologs shown to catalyze the transport of the plant hormone, auxin. Using the crystal structures of the multidrug exporters Sav1866 and MmABCB1 as templates, we have developed structural models of plant ABCB proteins with a common architecture. Comparisons of these structures identified kingdom-specific candidate substrate-binding regions within the translocation chamber formed by the transmembrane domains of ABCBs from the model plant Arabidopsis. These results suggest an early evolutionary divergence of plant and mammalian ABCBs. Validation of these models becomes a priority for efforts to elucidate ABCB function and manipulate this class of transporters to enhance plant productivity and quality.
Collapse
Affiliation(s)
- Aurélien Bailly
- Plant Biology, Department of Biology, University of FribourgFribourg, Switzerland
- Institute of Plant Biology, Zurich–Basel Plant Science Center, University of ZurichZurich, Switzerland
| | - Haibing Yang
- Department of Horticulture and Landscape Architecture, Purdue UniversityWest Lafayette, IN, USA
| | - Enrico Martinoia
- Institute of Plant Biology, Zurich–Basel Plant Science Center, University of ZurichZurich, Switzerland
| | - Markus Geisler
- Plant Biology, Department of Biology, University of FribourgFribourg, Switzerland
- Institute of Plant Biology, Zurich–Basel Plant Science Center, University of ZurichZurich, Switzerland
- *Correspondence: Markus Geisler, Plant Biology, Department of Biology, University of Fribourg, Chemin du Musée 10, CH-1700 Fribourg, Switzerland. e-mail:
| | - Angus S. Murphy
- Department of Horticulture and Landscape Architecture, Purdue UniversityWest Lafayette, IN, USA
| |
Collapse
|