1
|
Taneja N, Bersi MR, Rasmussen ML, Gama V, Merryman WD, Burnette DT. Inhibition of focal adhesion kinase increases myofibril viscosity in cardiac myocytes. Cytoskeleton (Hoboken) 2020; 77:342-350. [PMID: 32885903 DOI: 10.1002/cm.21632] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/25/2020] [Accepted: 08/28/2020] [Indexed: 11/06/2022]
Abstract
The coordinated generation of mechanical forces by cardiac myocytes is required for proper heart function. Myofibrils are the functional contractile units of force production within individual cardiac myocytes. At the molecular level, myosin motors form cross-bridges with actin filaments and use ATP to convert chemical energy into mechanical forces. The energetic efficiency of the cross-bridge cycle is influenced by the viscous damping of myofibril contraction. The viscoelastic response of myofibrils is an emergent property of their individual mechanical components. Previous studies have implicated titin-actin interactions, cell-ECM adhesion, and microtubules as regulators of the viscoelastic response of myofibrils. Here we probed the viscoelastic response of myofibrils using laser-assisted dissection. As a proof-of-concept, we found actomyosin contractility was required to endow myofibrils with their viscoelastic response, with blebbistatin treatment resulting in decreased myofibril tension and viscous damping. Focal adhesion kinase (FAK) is a key regulator of cell-ECM adhesion, microtubule stability, and myofibril assembly. We found inhibition of FAK signaling altered the viscoelastic properties of myofibrils. Specifically, inhibition of FAK resulted in increased viscous damping of myofibril retraction following laser ablation. This damping was not associated with acute changes in the electrophysiological properties of cardiac myocytes. These results implicate FAK as a regulator of mechanical properties of myofibrils.
Collapse
Affiliation(s)
- Nilay Taneja
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Matthew R Bersi
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Megan L Rasmussen
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Dylan T Burnette
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
2
|
Li Q, Wang Z. Involvement of FAK/P38 Signaling Pathways in Mediating the Enhanced Osteogenesis Induced by Nano-Graphene Oxide Modification on Titanium Implant Surface. Int J Nanomedicine 2020; 15:4659-4676. [PMID: 32636624 PMCID: PMC7335313 DOI: 10.2147/ijn.s245608] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/01/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Titanium implants are widely used in dental and orthopedic medicine. Nevertheless, there is limited osteoinductive capability of titanium leading to a poor or delayed osseointegration, which might cause the failure of the implant therapy. Therefore, appropriate modification on the titanium surface for promoting osseointegration of existing implants is still pursued. PURPOSE Graphene oxide (GO) is a promising candidate to perform implant surface biofunctionalization for modulating the interactions between implant surface and cells. So the objective of this study was to fabricate a bioactive GO-modified titanium implant surface with excellent osteoinductive potential and further investigate the underlying biological mechanisms. MATERIALS AND METHODS The large particle sandblasting and acid etching (SLA, commonly used in clinical practice) surface as a control group was first developed and then the nano-GO was deposited on the SLA surface via an ultrasonic atomization spraying technique to create the SLA/GO group. Their effects on rat bone marrow mesenchymal stem cells (BMSCs) responsive behaviors were assessed in vitro, and the underlying biological mechanisms were further systematically investigated. Moreover, the osteogenesis performance in vivo was also evaluated. RESULTS The results showed that GO coating was fabricated on the titanium substrates successfully, which endowed SLA surface with the improved hydrophilicity and protein adsorption capacity. Compared with the SLA surface, the GO-modified surface favored cell adhesion and spreading, and significantly improved cell proliferation and osteogenic differentiation of BMSCs in vitro. Furthermore, the FAK/P38 signaling pathways were proven to be involved in the enhanced osteogenic differentiation of BMSCs, accompanied by the upregulated expression of focal adhesion (vinculin) on the GO coated surface. The enhanced bone regeneration ability of GO-modified implants when inserted into rat femurs was also observed and confirmed that the GO coating induced accelerated osseointegration and osteogenesis in vivo. CONCLUSION GO modification on titanium implant surface has potential applications for achieving rapid bone-implant integration through the mediation of FAK/P38 signaling pathways.
Collapse
Affiliation(s)
- Qingfan Li
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, People’s Republic of China
- Department of Oral Implant, School of Stomatology, Hospital of Stomatology, Tongji University, Shanghai, People’s Republic of China
| | - Zuolin Wang
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, People’s Republic of China
- Department of Oral Implant, School of Stomatology, Hospital of Stomatology, Tongji University, Shanghai, People’s Republic of China
| |
Collapse
|
3
|
Huang MH, Liu PY, Wu SN. Characterization of Perturbing Actions by Verteporfin, a Benzoporphyrin Photosensitizer, on Membrane Ionic Currents. Front Chem 2019; 7:566. [PMID: 31508407 PMCID: PMC6714490 DOI: 10.3389/fchem.2019.00566] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/25/2019] [Indexed: 01/13/2023] Open
Abstract
Verteporfin (VP), a benzoporphyrin derivative, has been clinically tailored as a photosensitizer and recently known to suppress YAP-TEAD complex accompanied by suppression of the growth in an array of neoplastic cells. However, the detailed information is little available regarding possible modifications of it and its related compounds on transmembrane ionic currents, despite its growing use in clinical settings. In this study, from whole cell recordings, VP (0.3-100 μM) increased the amplitude of Ca2+-activated K+ currents (I K(Ca)) in pituitary tumor (GH3) cells in a concentration-dependent manner with an EC50 value of 2.4 μM. VP-stimulated I K(Ca) in these cells was suppressed by further addition of either paxilline, iberiotoxin, or dithiothreitol, but not by that of tobultamide or TRAM-39. VP at a concentration of 10 μM mildly suppressed the amplitude of delayed-rectifier K+ current; however, it had minimal effects on M-type K+ current. In cell-attached current recordings, addition of VP to the recording medium enhanced the activity of large-conductance Ca2+-activated K+ (BKCa) channels. In the presence of VP, additional illumination with light intensity of 5.5 mW/cm2 raised the probability of BKCa-channel openings further. Addition of VP decreased the peak amplitude of L-type Ca2+ current together with slowed inactivation time course of the current; however, it failed to modify voltage-gated Na+ current. Illumination of GH3 cells in continued presence of VP also induced a non-selective cation current. Additionally, VP increased the activity of BKCa channels in human 13-06-MG glioma cells with an EC50 value of 1.9 μM. Therefore, the effects of VP on ionic currents described herein tend to be upstream of its inhibition of YAP-TEAD complex and they are conceivably likely to contribute to the underlying mechanisms through which it and its structurally similar compounds effect the modifications in functional activities of pituitary or glial neoplastic cells, if the in vivo findings occur.
Collapse
Affiliation(s)
- Mei-Han Huang
- College of Medical and Health Sciences, Fooyin University, Kaohsiung City, Taiwan
| | - Ping-Yen Liu
- Division of Cardiovascular Medicine, National Cheng Kung University Medical College, Tainan City, Taiwan
| | - Sheng-Nan Wu
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan City, Taiwan.,Department of Physiology, National Cheng Kung University Medical College, Tainan City, Taiwan
| |
Collapse
|
4
|
Zhang X, Li H, Liu J, Wang H, Sun W, Lin K, Wang X, Shen SG. Amorphous carbon modification on implant surface: a general strategy to enhance osteogenic differentiation for diverse biomaterials via FAK/ERK1/2 signaling pathways. J Mater Chem B 2019; 7:2518-2533. [DOI: 10.1039/c8tb02850h] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Amorphous carbon coatings enhance osteogenic differentiation via FAK/ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Xinran Zhang
- Department of Oral and Cranio-Maxillofacial Science
- Shanghai Ninth People's Hospital
- College of Stomatology
- Shanghai Jiao Tong University School of Medicine
- National Clinical Research Center for Oral Diseases
| | - Haotian Li
- Department of Spine Surgery
- Tongji Hospital
- Tongji University School of Medicine
- Shanghai 200065
- China
| | - Jiaqiang Liu
- Department of Oral and Cranio-Maxillofacial Science
- Shanghai Ninth People's Hospital
- College of Stomatology
- Shanghai Jiao Tong University School of Medicine
- National Clinical Research Center for Oral Diseases
| | - Hui Wang
- School & Hospital of Stomatology
- Tongji University
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration
- Shanghai 200072
- China
| | - Wenjun Sun
- School & Hospital of Stomatology
- Tongji University
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration
- Shanghai 200072
- China
| | - Kaili Lin
- Department of Oral and Cranio-Maxillofacial Science
- Shanghai Ninth People's Hospital
- College of Stomatology
- Shanghai Jiao Tong University School of Medicine
- National Clinical Research Center for Oral Diseases
| | - Xudong Wang
- Department of Oral and Cranio-Maxillofacial Science
- Shanghai Ninth People's Hospital
- College of Stomatology
- Shanghai Jiao Tong University School of Medicine
- National Clinical Research Center for Oral Diseases
| | - Steve Guofang Shen
- Department of Oral and Cranio-Maxillofacial Science
- Shanghai Ninth People's Hospital
- College of Stomatology
- Shanghai Jiao Tong University School of Medicine
- National Clinical Research Center for Oral Diseases
| |
Collapse
|
5
|
Haynes JM, Selby JN, Vandekolk TH, Abad IPL, Ho JK, Lieuw WL, Leach K, Savige J, Saini S, Fisher CL, Ricardo SD. Induced Pluripotent Stem Cell-Derived Podocyte-Like Cells as Models for Assessing Mechanisms Underlying Heritable Disease Phenotype: Initial Studies Using Two Alport Syndrome Patient Lines Indicate Impaired Potassium Channel Activity. J Pharmacol Exp Ther 2018; 367:335-347. [PMID: 30104322 DOI: 10.1124/jpet.118.250142] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/26/2018] [Indexed: 12/22/2022] Open
Abstract
Renal podocyte survival depends upon the dynamic regulation of a complex cell architecture that links the glomerular basement membrane to integrins, ion channels, and receptors. Alport syndrome is a heritable chronic kidney disease where mutations in α3, α4, or α5 collagen genes promote podocyte death. In rodent models of renal failure, activation of the calcium-sensing receptor (CaSR) can protect podocytes from stress-related death. In this study, we assessed CaSR function in podocyte-like cells derived from induced-pluripotent stem cells from two patients with Alport Syndrome (AS1 & AS2) and a renal disease free individual [normal human mesangial cell (NHMC)], as well as a human immortalized podocyte-like (HIP) cell line. Extracellular calcium elicited concentration-dependent elevations of intracellular calcium in all podocyte-like cells. NHMC and HIP, but not AS1 or AS2 podocyte-like cells, also showed acute reductions in intracellular calcium prior to elevation. In NHMC podocyte-like cells this acute reduction was blocked by the large-conductance potassium channel (KCNMA1) inhibitors iberiotoxin (10 nM) and tetraethylammonium (5 mM), as well as the focal adhesion kinase inhibitor PF562271 (N-methyl-N-(3-((2-(2-oxo-2,3-dihydro-1H-indol-5-ylamino)-5-trifluoromethyl-pyrimidin-4-ylamino)-methyl)-pyridin-2-yl)-methanesulfonamide, 10 nM). Quantitative polymerase chain reaction (qPCR) and immunolabeling showed the presence of KCNMA1 transcript and protein in all podocyte-like cells tested. Cultivation of AS1 podocytes on decellularized plates of NHMC podocyte-like cells partially restored acute reductions in intracellular calcium in response to extracellular calcium. We conclude that the AS patient-derived podocyte-like cells used in this study showed dysfunctional integrin signaling and potassium channel function, which may contribute to podocyte death seen in Alport syndrome.
Collapse
Affiliation(s)
- John M Haynes
- Monash Institute of Pharmaceutical Sciences (J.M.H., J.N.S., T.H.V., I.P.L.A., J.K.H., W.-L.L., K.L.) and Department of Anatomy and Developmental Biology (S.S., C.L.F., S.D.R.), Monash University, Victoria, Australia; and Department of Medicine, Royal Melbourne Hospital, Victoria, Australia (J.S.)
| | - James N Selby
- Monash Institute of Pharmaceutical Sciences (J.M.H., J.N.S., T.H.V., I.P.L.A., J.K.H., W.-L.L., K.L.) and Department of Anatomy and Developmental Biology (S.S., C.L.F., S.D.R.), Monash University, Victoria, Australia; and Department of Medicine, Royal Melbourne Hospital, Victoria, Australia (J.S.)
| | - Teresa H Vandekolk
- Monash Institute of Pharmaceutical Sciences (J.M.H., J.N.S., T.H.V., I.P.L.A., J.K.H., W.-L.L., K.L.) and Department of Anatomy and Developmental Biology (S.S., C.L.F., S.D.R.), Monash University, Victoria, Australia; and Department of Medicine, Royal Melbourne Hospital, Victoria, Australia (J.S.)
| | - Isaiah P L Abad
- Monash Institute of Pharmaceutical Sciences (J.M.H., J.N.S., T.H.V., I.P.L.A., J.K.H., W.-L.L., K.L.) and Department of Anatomy and Developmental Biology (S.S., C.L.F., S.D.R.), Monash University, Victoria, Australia; and Department of Medicine, Royal Melbourne Hospital, Victoria, Australia (J.S.)
| | - Joan K Ho
- Monash Institute of Pharmaceutical Sciences (J.M.H., J.N.S., T.H.V., I.P.L.A., J.K.H., W.-L.L., K.L.) and Department of Anatomy and Developmental Biology (S.S., C.L.F., S.D.R.), Monash University, Victoria, Australia; and Department of Medicine, Royal Melbourne Hospital, Victoria, Australia (J.S.)
| | - Wai-Ling Lieuw
- Monash Institute of Pharmaceutical Sciences (J.M.H., J.N.S., T.H.V., I.P.L.A., J.K.H., W.-L.L., K.L.) and Department of Anatomy and Developmental Biology (S.S., C.L.F., S.D.R.), Monash University, Victoria, Australia; and Department of Medicine, Royal Melbourne Hospital, Victoria, Australia (J.S.)
| | - Katie Leach
- Monash Institute of Pharmaceutical Sciences (J.M.H., J.N.S., T.H.V., I.P.L.A., J.K.H., W.-L.L., K.L.) and Department of Anatomy and Developmental Biology (S.S., C.L.F., S.D.R.), Monash University, Victoria, Australia; and Department of Medicine, Royal Melbourne Hospital, Victoria, Australia (J.S.)
| | - Judith Savige
- Monash Institute of Pharmaceutical Sciences (J.M.H., J.N.S., T.H.V., I.P.L.A., J.K.H., W.-L.L., K.L.) and Department of Anatomy and Developmental Biology (S.S., C.L.F., S.D.R.), Monash University, Victoria, Australia; and Department of Medicine, Royal Melbourne Hospital, Victoria, Australia (J.S.)
| | - Sheetal Saini
- Monash Institute of Pharmaceutical Sciences (J.M.H., J.N.S., T.H.V., I.P.L.A., J.K.H., W.-L.L., K.L.) and Department of Anatomy and Developmental Biology (S.S., C.L.F., S.D.R.), Monash University, Victoria, Australia; and Department of Medicine, Royal Melbourne Hospital, Victoria, Australia (J.S.)
| | - Craig L Fisher
- Monash Institute of Pharmaceutical Sciences (J.M.H., J.N.S., T.H.V., I.P.L.A., J.K.H., W.-L.L., K.L.) and Department of Anatomy and Developmental Biology (S.S., C.L.F., S.D.R.), Monash University, Victoria, Australia; and Department of Medicine, Royal Melbourne Hospital, Victoria, Australia (J.S.)
| | - Sharon D Ricardo
- Monash Institute of Pharmaceutical Sciences (J.M.H., J.N.S., T.H.V., I.P.L.A., J.K.H., W.-L.L., K.L.) and Department of Anatomy and Developmental Biology (S.S., C.L.F., S.D.R.), Monash University, Victoria, Australia; and Department of Medicine, Royal Melbourne Hospital, Victoria, Australia (J.S.)
| |
Collapse
|
6
|
Qu M, Liu Z, Zhao D, Wang C, Zhang J, Tang Z, Liu K, Shu X, Yuan H, Ma X. Design, synthesis and biological evaluation of sulfonamide-substituted diphenylpyrimidine derivatives (Sul-DPPYs) as potent focal adhesion kinase (FAK) inhibitors with antitumor activity. Bioorg Med Chem 2017; 25:3989-3996. [PMID: 28576633 DOI: 10.1016/j.bmc.2017.05.044] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 05/17/2017] [Accepted: 05/20/2017] [Indexed: 10/19/2022]
Abstract
A class of sulfonamide-substituted diphenylpyrimidines (Sul-DPPYs) were synthesized to improve activity against the focal adhesion kinase (FAK). Most of these new Sul-DPPYs displayed moderate activity against the FAK enzyme with IC50 values of less than 100nM; regardless, they could effectively inhibit several classes of refractory cancer cell lines with IC50 values of less than 10µM, including the pancreatic cancer cell lines (AsPC-1, Panc-1 and BxPC-3), the NSCLC-resistant H1975 cell line, and the B lymphocyte cell line (Ramos cells). Results of flow cytometry indicated that inhibitor 7e promoted apoptosis of pancreatic cancer cells in a dose-dependent manner. In addition, it almost completely induced the apoptosis at a concentration of 10µM. Compound 7e may be selected as a potent FAK inhibitor for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Menghua Qu
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Zhihao Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Dan Zhao
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Changyuan Wang
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Jianbin Zhang
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Zeyao Tang
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Kexin Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Xiaohong Shu
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China.
| | - Hong Yuan
- Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, PR China.
| | - Xiaodong Ma
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China.
| |
Collapse
|
7
|
Zhao X, Sun W, Puszyk WM, Wallet S, Hochwald S, Robertson K, Liu C. Focal adhesion kinase inhibitor PF573228 and death receptor 5 agonist lexatumumab synergistically induce apoptosis in pancreatic carcinoma. Tumour Biol 2017; 39:1010428317699120. [PMID: 28459212 DOI: 10.1177/1010428317699120] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
Pancreatic cancer has one of the lowest survival rates of all cancers. The mechanism underlying chemo-resistance of pancreatic cancer is not well understood. Our previous article reported that small molecule YM155 induced apoptosis in pancreatic cancer cells via activation of death receptor 5. In this study, we aim to continuously address death receptor 5-mediated apoptosis in chemo-resistant pancreatic carcinoma. We found that in comparison to paired pancreatic cancer tissues and adjacent normal tissues, five of the six cancer tissues had downregulated death receptor 5 and upregulated Bcl-xL. Mono treatment with lexatumumab was not sufficient to induce apoptosis in pancreatic cancer cells, whereas focal adhesion kinase inhibitor PF573228 significantly sensitized lexatumumab-induced apoptosis. Western blotting analysis revealed that lexatumumab and PF573228 combination treatment increased death receptor 5 but decreased Bcl-xL expression. Interestingly, pre-treatment with Bcl-xL inhibitor ABT263 reversed the insensitivity of panc-1 cells to lexatumumab or PF573228-induced apoptosis. Specific small interfering RNA-mediated gene silencing of Bcl-xL effectively sensitized pancreatic cancer cells to lexatumumab or PF573228-induced apoptosis. Furthermore, lexatumumab and PF573228 combination was shown to exhibit significant xenograft pancreatic tumor growth inhibition in SCID mice. Our data provide fundamental evidence to support the notion that lexatumumab and PF573228 co-treatment could be a potentially effective regime for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Xiangxuan Zhao
- 1 Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
- 2 Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Wei Sun
- 1 Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - William M Puszyk
- 2 Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Shannon Wallet
- 3 Department of Periodontology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - Steve Hochwald
- 4 Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Keith Robertson
- 5 Departments of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Chen Liu
- 2 Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
8
|
Wu SN, Chen HZ, Chou YH, Huang YM, Lo YC. Inhibitory actions by ibandronate sodium, a nitrogen-containing bisphosphonate, on calcium-activated potassium channels in Madin-Darby canine kidney cells. Toxicol Rep 2015; 2:1182-1193. [PMID: 28962460 PMCID: PMC5598360 DOI: 10.1016/j.toxrep.2015.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/23/2015] [Accepted: 08/25/2015] [Indexed: 12/15/2022] Open
Abstract
The nitrogen-containing bisphosphonates used for management of the patients with osteoporosis were reported to influence the function of renal tubular cells. However, how nitrogen-containing bisphosphates exert any effects on ion currents remains controversial. The effects of ibandronate (Iban), a nitrogen-containing bisphosphonate, on ionic channels, including two types of Ca2+-activated K+ (KCa) channels, namely, large-conductance KCa (BKCa) and intermediate-conductance KCa (IKCa) channels, were investigated in Madin–Darby canine kidney (MDCK) cells. In whole-cell current recordings, Iban suppressed the amplitude of voltage-gated K+ current elicited by long ramp pulse. Addition of Iban caused a reduction of BKCa channels accompanied by a right shift in the activation curve of BKCa channels, despite no change in single-channel conductance. Ca2+ sensitivity of these channels was modified in the presence of this compound; however, the magnitude of Iban-mediated decrease in BKCa-channel activity under membrane stretch with different negative pressure remained unchanged. Iban suppressed the probability of BKCa-channel openings linked primarily to a shortening in the slow component of mean open time in these channels. The dissociation constant needed for Iban-mediated suppression of mean open time in MDCK cells was 12.2 μM. Additionally, cell exposure to Iban suppressed the activity of IKCa channels, and DC-EBIO or 9-phenanthrol effectively reversed its suppression. Under current-clamp configuration, Iban depolarized the cells and DC-EBIO or PF573228 reversed its depolarizing effect. Taken together, the inhibitory action of Iban on KCa-channel activity may contribute to the underlying mechanism of pharmacological or toxicological actions of Iban and its structurally similar bisphosphonates on renal tubular cells occurring in vivo.
Collapse
Key Words
- BKCa channel, large-conductance Ca2+-activated K+ channel
- DC-EBIO, 5,6-dichloro-1-ethyl-1,3-dihydro-2H-benzimidazol-2-one
- DMEM, Dulbecco’s modified Eagle’s medium
- FBS, fetal bovine serum
- H2S, hydrogen sulfide
- IK, voltage-gated K+ current
- IKCa channel, intermediate-conductance Ca2+-activated K+ channel
- Iban, ibandronate sodium
- Ibandronate
- Intermediate-conductance Ca2+-activated K+ channel
- I–V, current–voltage
- KCa channel, Ca2+-activated K+ channel
- KD, dissociation constant
- Large-conductance Ca2+-activated K+ channel
- MDCK cell
- MDCK cell, Madin–Darby canine kidney cell,NaHS, sodium hydrosulphide
- Membrane potential
- PF573228, 3,4-dihydro-6-[[4-[[[3-(methylsulfonyl)phenyl]methyl]amino]-5(trifluoromethyl)-2-pyrimidinyl]amino]-2(1H)-quinolinone
- SEM, standard error of the mean
- TRAM-34, 1-((2-chloropheny) (diphenyl)methyl)-1H-pyrazole
- [Ca2+]i, intracellular Ca2+ concentration
Collapse
Affiliation(s)
- Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan City, Taiwan
| | - Hui-Zhen Chen
- Department of Physiology, National Cheng Kung University Medical College, Tainan City, Taiwan
| | - Yu-Hung Chou
- School of Medicine, Mayne Medical School, University of Queensland, Brisbane, Queensland, Australia
| | - Yan-Ming Huang
- Department of Physiology, National Cheng Kung University Medical College, Tainan City, Taiwan
| | - Yi-Ching Lo
- Department of Pharmacology, Kaohsiung Medical University, Kaohsiung City, Taiwan
| |
Collapse
|
9
|
Colinas O, Moreno-Domínguez A, Zhu HL, Walsh EJ, Pérez-García MT, Walsh MP, Cole WC. α5-Integrin-mediated cellular signaling contributes to the myogenic response of cerebral resistance arteries. Biochem Pharmacol 2015; 97:281-91. [PMID: 26278977 DOI: 10.1016/j.bcp.2015.08.088] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/10/2015] [Indexed: 12/24/2022]
Abstract
The myogenic response of resistance arterioles and small arteries involving constriction in response to intraluminal pressure elevation and dilation on pressure reduction is fundamental to local blood flow regulation in the microcirculation. Integrins have garnered considerable attention in the context of initiating the myogenic response, but evidence indicative of mechanotransduction by integrin adhesions, for example established changes in tyrosine phosphorylation of key adhesion proteins, has not been obtained to substantiate this interpretation. Here, we evaluated the role of integrin adhesions and associated cellular signaling in the rat cerebral arterial myogenic response using function-blocking antibodies against α5β1-integrins, pharmacological inhibitors of focal adhesion kinase (FAK) and Src family kinase (SFK), an ultra-high-sensitivity western blotting technique, site-specific phosphoprotein antibodies to quantify adhesion and contractile filament protein phosphorylation, and differential centrifugation to determine G-actin levels in rat cerebral arteries at varied intraluminal pressures. Pressure-dependent increases in the levels of phosphorylation of FAK (FAK-Y397, Y576/Y577), SFK (SFK-Y416; Y527 phosphorylation was reduced), vinculin-Y1065, paxillin-Y118 and phosphoinositide-specific phospholipase C-γ1 (PLCγ1)-Y783 were detected. Treatment with α5-integrin function-blocking antibodies, FAK inhibitor FI-14 or SFK inhibitor SU6656 suppressed the changes in adhesion protein phosphorylation, and prevented pressure-dependent phosphorylation of the myosin targeting subunit of myosin light chain phosphatase (MYPT1) at T855 and 20kDa myosin regulatory light chains (LC20) at S19, as well as actin polymerization that are necessary for myogenic constriction. We conclude that mechanotransduction by integrin adhesions and subsequent cellular signaling play a fundamental role in the cerebral arterial myogenic response.
Collapse
Affiliation(s)
- Olaia Colinas
- Smooth Muscle Research Group, Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Libin Cardiovascular Institute, University of Calgary, Alberta, Canada.
| | - Alejandro Moreno-Domínguez
- Smooth Muscle Research Group, Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Libin Cardiovascular Institute, University of Calgary, Alberta, Canada.
| | - Hai-Lei Zhu
- Smooth Muscle Research Group, Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Libin Cardiovascular Institute, University of Calgary, Alberta, Canada.
| | - Emma J Walsh
- Smooth Muscle Research Group, Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Libin Cardiovascular Institute, University of Calgary, Alberta, Canada.
| | - M Teresa Pérez-García
- Department of Physiology, Instituto de Biología y Genética Molecular, University of Valladolid, Valladolid, Spain.
| | - Michael P Walsh
- Smooth Muscle Research Group, Department of Biochemistry and Molecular Biology, Hotchkiss Brain Institute and Libin Cardiovascular Institute, University of Calgary, Alberta, Canada.
| | - William C Cole
- Smooth Muscle Research Group, Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Libin Cardiovascular Institute, University of Calgary, Alberta, Canada.
| |
Collapse
|
10
|
Wu SN, Huang YM, Liao YK. Effects of ibandronate sodium, a nitrogen-containing bisphosphonate, on intermediate-conductance calcium-activated potassium channels in osteoclast precursor cells (RAW 264.7). J Membr Biol 2015; 248:103-115. [PMID: 25362532 DOI: 10.1007/s00232-014-9747-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 10/23/2014] [Indexed: 01/08/2023]
Abstract
Ibanonate sodium (Iban), a nitrogen-containing bisphosphonate, is recognized to reduce skeletal complications through an inhibition of osteoclast-mediated bone resorption. However, how this drug interacts with ion channels in osteoclasts and creates anti-osteoclastic activity remains largely unclear. In this study, we investigated the possible effects of Iban and other related compounds on ionic currents in the osteoclast precursor RAW 264.7 cells. Iban suppressed the amplitude of whole-cell K(+) currents (I K) in a concentration-dependent manner with an IC50 value of 28.9 μM. The I K amplitude was sensitive to block by TRAM-34 and Iban-mediated inhibition of I K was reversed by further addition of DCEBIO, an activator of intermediate-conductance Ca(2+)-activated K(+) (IKCa) channels. Intracellular dialysis with Iban diminished I K amplitude and further addition of ionomycin reversed its inhibition. In 17β-estradiol-treated cells, Iban-mediated inhibition of I K remained effective. In cell-attached current recordings, Iban applied to bath did not modify single-channel conductance of IKCa channels; however, it did reduce channel activity. Iban-induced inhibition of IKCa channels was voltage-dependent. As IKCa-channel activity was suppressed by KN-93, subsequent addition of Iban did not further decrease the channel open probability. Iban could not exert any effect on inwardly rectifying K(+) current in RAW 264.7 cells. Under current-clamp recordings, Iban depolarized the membrane of RAW 264.7 cells and DCEBIO reversed Iban-induced depolarization. Iban also suppressed lipopolysaccharide-stimulated migration of RAW 264.7 cells in a concentration-dependent manner. Therefore, the inhibition by Iban of IKCa channels would be an important mechanism underlying its actions on the functional activity of osteoclasts occurring in vivo.
Collapse
Affiliation(s)
- Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, No. 1 University Road, Tainan City, 70101, Taiwan,
| | | | | |
Collapse
|
11
|
Hsu HT, Tseng YT, Lo YC, Wu SN. Ability of naringenin, a bioflavonoid, to activate M-type potassium current in motor neuron-like cells and to increase BKCa-channel activity in HEK293T cells transfected with α-hSlo subunit. BMC Neurosci 2014; 15:135. [PMID: 25539574 PMCID: PMC4288500 DOI: 10.1186/s12868-014-0135-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 12/11/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Naringenin (NGEN) is a citrus bioflavonoid known to have beneficial health properties; however, the ionic mechanism of its actions remains largely unclear. In this study, we attempted to evaluate the possible effects of NGEN on K(+) currents in NSC-34 neuronal cells and in HEK293T cells expressing α-hSlo. RESULTS NGEN increased M-type K(+) current (I(K(M))) in a concentration-dependent manner with an EC50 value of 9.8 μM in NSC-34 cells. NGEN shifted the activation curve of I(K(M)) conductance to the more negative potentials. In cell-attached recordings, NGEN or flupirtine enhanced the activity of M-type K(+) (K(M)) channels with no changes in single-channel amplitude. NGEN (10 μM) had minimal effect on erg-mediated K(+) currents. Under cell-attached voltage-clamp recordings, NGEN decreased the frequency of spontaneous action currents and further application of linopirdine can reverse NGEN-induced inhibition of firing. In HEK293T cells expressing α-hSlo, this compound increased the amplitude of Ca(2+)-activated K(+) current (I(K(Ca))). Under inside-out recordings, NGEN applied to the intracellular side of the detached patch enhanced the activity of large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels. Moreover, from the study of a modeled neuron, burst firing of simulated action potentials (APs) was reduced in the presence of the increased conductances of both K(M) and K(Ca) channels. Fast-slow analysis of AP bursting from this model also revealed that as the conductances of both K(M) and BK(Ca) channels were increased by two-fold, the voltage nullcline was shifted in an upward direction accompanied by the compression of burst trajectory. CONCLUSIONS The present results demonstrate that activation of both K(M) and BK(Ca) channels caused by NGEN might combine to influence neuronal activity if similar channels were functionally co-expressed in central neurons in vivo.
Collapse
Affiliation(s)
- Hung-Te Hsu
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Department of Anesthesia, Kaohsiung Medical University Hospital, Kaohsiung City, 80708, Taiwan.
| | - Yu-Ting Tseng
- Department of Pharmacology, School of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Graduate Institute of Natural Products, School of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Yi-Ching Lo
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Department of Pharmacology, School of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Graduate Institute of Natural Products, School of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan City, 70101, Taiwan.
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan City, 70101, Taiwan.
| |
Collapse
|
12
|
Schickling BM, England SK, Aykin-Burns N, Norian LA, Leslie KK, Frieden-Korovkina VP. BKCa channel inhibitor modulates the tumorigenic ability of hormone-independent breast cancer cells via the Wnt pathway. Oncol Rep 2014; 33:533-8. [PMID: 25422049 PMCID: PMC4306270 DOI: 10.3892/or.2014.3617] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/12/2014] [Indexed: 11/05/2022] Open
Abstract
In breast cancers, the large conductance Ca2+ and voltage sensitive K+ (BKCa) channels have been hypothesized to function as oncoproteins, yet it remains unclear how inhibition of channel activity impacts oncogenesis. We demonstrated herein that iberiotoxin (IbTX), an inhibitor of BKCa channels, differentially modulated the in vitro tumorigenic activities of hormone-independent breast cancer cells. Specifically, in HER-2/neu-overexpressing UACC893 cells and triple‑negative MDA-MB-231 cells, IbTX selectively attenuated anchorage-independent growth with concomitant downregulation of β-catenin as well as total and phosphorylated Akt and HER-2/neu. By contrast, HER-2/neu-overexpressing SK-BR-3 cells were insensitive to IbTX. Molecular analyses showed an absence of β-catenin and a dose-dependent upregulation of total and phosphorylated Akt and HER-2/neu in these cells. Taken together, these studies identify β-catenin as a putative modulator of the inhibitory actions of IbTX in sensitive breast cancer cells.
Collapse
Affiliation(s)
| | - Sarah K England
- Division of Basic Science Research, Washington University, St. Louis, MO, USA
| | - Nukhet Aykin-Burns
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lyse A Norian
- Department of Urology, University of Iowa, Iowa City, IA, USA
| | - Kimberly K Leslie
- Department of Obstetrics and Gynecology, University of Iowa, Iowa City, IA, USA
| | | |
Collapse
|
13
|
Kunit T, Gratzke C, Schreiber A, Strittmatter F, Waidelich R, Rutz B, Loidl W, Andersson KE, Stief CG, Hennenberg M. Inhibition of smooth muscle force generation by focal adhesion kinase inhibitors in the hyperplastic human prostate. Am J Physiol Renal Physiol 2014; 307:F823-32. [DOI: 10.1152/ajprenal.00011.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Smooth muscle contraction may be critical for lower urinary tract symptoms (LUTS) in patients with benign prostate hyperplasia and requires stable anchorage of the cytoskeleton to the cell membrane. These connections are regulated by focal adhesion kinase (FAK). Here, we addressed the involvement of FAK in the regulation of smooth muscle contraction in hyperplastic human prostate tissues. Prostate tissues were obtained from radical prostatectomy. Expression of FAK and focal adhesion proteins was assessed by Western blot analysis and immunohistochemical stainings. Effects of the FAK inhibitors PF-573228 and Y-11 on contraction of prostate strips were examined in the organ bath. Expression of FAK and focal adhesion proteins (integrin-5α, paxilin, and c-Src) was detected by Western blot analysis in prostate samples. By double immunofluorescence staining with calponin and pan-cytokeratin, expression of FAK was observed in stromal and epithelial cells. Immunoreactivity for FAK colocalized with integrin-5α, paxilin, talin, and c-Src. Stimulation of prostate tissues with the α1-adrenergic agonist phenylephrine increased the phosphorylation state of FAK at Tyr397 and Tyr925 with different kinetics, which was blocked by the α1-adrenoceptor antagonist tamsulosin. Norepinephrine and phenylephrine induced concentration-dependent contractions of prostate strips. Both FAK inhibitors PF-573228 and Y-11 significantly inhibited norepinephrine- and phenylephrine-induced contractions. Finally, PF-573228 and Y-11 inhibited contractions induced by electric field stimulation, which was significant at the highest frequency. In conclusion, α1-adrenergic smooth muscle contraction or its regulation involves FAK in the human prostate. Consequently, FAK may be involved in the pathophysiology of LUTS and in current or future LUTS therapies.
Collapse
Affiliation(s)
- Thomas Kunit
- Department of Urology, Ludwig-Maximilians University, Munich, Germany
- Krankenhaus der Barmherzigen Schwestern Linz, Linz, Austria; and
| | - Christian Gratzke
- Department of Urology, Ludwig-Maximilians University, Munich, Germany
| | - Andrea Schreiber
- Department of Urology, Ludwig-Maximilians University, Munich, Germany
| | | | | | - Beata Rutz
- Department of Urology, Ludwig-Maximilians University, Munich, Germany
| | - Wolfgang Loidl
- Krankenhaus der Barmherzigen Schwestern Linz, Linz, Austria; and
| | - Karl-Erik Andersson
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | | | - Martin Hennenberg
- Department of Urology, Ludwig-Maximilians University, Munich, Germany
| |
Collapse
|
14
|
Cheung So E, Lo YC, Chen LT, Kao CA, Wu SN. High effectiveness of triptolide, an active diterpenoid triepoxide, in suppressing Kir-channel currents from human glioma cells. Eur J Pharmacol 2014; 738:332-341. [DOI: 10.1016/j.ejphar.2014.05.059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 05/23/2014] [Accepted: 05/23/2014] [Indexed: 11/17/2022]
|
15
|
Hosseinzadeh Z, Almilaji A, Honisch S, Pakladok T, Liu G, Bhavsar SK, Ruth P, Shumilina E, Lang F. Upregulation of the large conductance voltage- and Ca2+-activated K+ channels by Janus kinase 2. Am J Physiol Cell Physiol 2014; 306:C1041-9. [PMID: 24696148 DOI: 10.1152/ajpcell.00209.2013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The iberiotoxin-sensitive large conductance voltage- and Ca(2+)-activated potassium (BK) channels (maxi-K(+)-channels) hyperpolarize the cell membrane thus supporting Ca(2+) entry through Ca(2+)-release activated Ca(2+) channels. Janus kinase-2 (JAK2) has been identified as novel regulator of ion transport. To explore whether JAK2 participates in the regulation of BK channels, cRNA encoding Ca(2+)-insensitive BK channels (BK(M513I+Δ899-903)) was injected into Xenopus oocytes with or without cRNA encoding wild-type JAK2, gain-of-function (V617F)JAK2, or inactive (K882E)JAK2. K(+) conductance was determined by dual electrode voltage clamp and BK-channel protein abundance by confocal microscopy. In A204 alveolar rhabdomyosarcoma cells, iberiotoxin-sensitive K(+) current was determined utilizing whole cell patch clamp. A204 cells were further transfected with JAK2 and BK-channel transcript, and protein abundance was quantified by RT-PCR and Western blotting, respectively. As a result, the K(+) current in BK(M513I+Δ899-903)-expressing oocytes was significantly increased following coexpression of JAK2 or (V617F)JAK2 but not (K882E)JAK2. Coexpression of the BK channel with (V617F)JAK2 but not (K882E)JAK2 enhanced BK-channel protein abundance in the oocyte cell membrane. Exposure of BK-channel and (V617F)JAK2-expressing oocytes to the JAK2 inhibitor AG490 (40 μM) significantly decreased K(+) current. Inhibition of channel insertion by brefeldin A (5 μM) decreased the K(+) current to a similar extent in oocytes expressing the BK channel alone and in oocytes expressing the BK channel and (V617F)JAK2. The iberiotoxin (50 nM)-sensitive K(+) current in rhabdomyosarcoma cells was significantly decreased by AG490 pretreatment (40 μM, 12 h). Moreover, overexpression of JAK2 in A204 cells significantly enhanced BK channel mRNA and protein abundance. In conclusion, JAK2 upregulates BK channels by increasing channel protein abundance in the cell membrane.
Collapse
Affiliation(s)
| | - Ahmad Almilaji
- Department of Physiology, University of Tübingen, Tübingen, Germany; and
| | - Sabina Honisch
- Department of Physiology, University of Tübingen, Tübingen, Germany; and
| | - Tatsiana Pakladok
- Department of Physiology, University of Tübingen, Tübingen, Germany; and
| | - GuoXing Liu
- Department of Physiology, University of Tübingen, Tübingen, Germany; and
| | - Shefalee K Bhavsar
- Department of Physiology, University of Tübingen, Tübingen, Germany; and
| | - Peter Ruth
- Institute of Pharmacy, Department of Pharmacology and Toxicology, University of Tübingen, Tübingen, Germany
| | | | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany; and
| |
Collapse
|
16
|
Effects of ketamine and its metabolites on ion currents in differentiated hippocampal H19-7 neuronal cells and in HEK293T cells transfected with α-hslo subunit. Neurotoxicology 2013; 33:1058-66. [PMID: 23227486 DOI: 10.1016/j.neuro.2012.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Ketamine (KT), a dissociative anesthetic, is known to induce schizophrenia-like psychosis. The percentage of KT abuse has recently grown fast despite KT being a controlled drug. The mechanism of KT actions is related to the inhibition of NMDA receptors. Whether KT produces other effects on ion currents in hippocampal neurons remains unclear. In this study, we attempted to evaluate the possible effects of KT and other related compounds on ion currents in hippocampal neuron-derived H19-7 cells. This drug exerted an inhibitory effect on Ca(2+)-activated K(+) current (IK(Ca)) in these cells with an IC(50) value of 274 μM. Pimaric acid (30 μM) or abietic acid (30 μM), known to stimulate large-conductance Ca(2+)-activated K(+) channels, reversed KT-induced inhibition of I(K)(Ca). In HEK293T cells expressing a-humans low poke, KT-induced inhibition of I(K)(Ca) still existed. Dehydronorketamine (300 μM) had little or no effect on the IK(Ca) amplitude, while norketamine (300 μM) slightly but significantly suppressed it. In inside–out configuration, KT applied to the intracellular face of the membrane did not alter single channel conductance of large-conductance Ca(2+)-activated K(+) (BKCa) channels; however, it did significantly reduce the probability of channel openings. Addition of KT was effective in depressing the peak amplitude of voltage-gated Na(+) current. Moreover, the presence of KT was noted to enhance the amplitude of membrane electroporation-induced inward currents (IMEP) in differentiated H19-7 cells. KT-stimulated IMEP was reversed by further application of LaCl(3) (100 μM), but not by NMDA (30 μM). The modulations by this compound of ion channels may contribute to the underlying mechanisms through which KT and its metabolites influence the electrical behavior of hippocampal neurons if similar findings occur in vivo.
Collapse
|