1
|
Barenco-Marins TS, Seara FAC, Ponte CG, Nascimento JHM. Pulmonary Circulation Under Pressure: Pathophysiological and Therapeutic Implications of BK Channel. Cardiovasc Drugs Ther 2023:10.1007/s10557-023-07503-7. [PMID: 37624526 DOI: 10.1007/s10557-023-07503-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/10/2023] [Indexed: 08/26/2023]
Abstract
The large-conductance Ca2+-activated K+ (BK) channel is widely expressed in the pulmonary blood vessels and plays a significant role in regulating pulmonary vascular tonus. It opens under membrane depolarization, increased intracellular Ca+2 concentration, and chronic hypoxia, resulting in massive K+ efflux, membrane hyperpolarization, decreased L-type Ca+2 channel opening, and smooth muscle relaxation. Several reports have demonstrated an association between BK channel dysfunction and pulmonary hypertension (PH) development. Decreased BK channel subunit expression and impaired regulation by paracrine hormones result in decreased BK channel opening, increased pulmonary vascular resistance, and pulmonary arterial pressure being the cornerstone of PH. The resulting right ventricular pressure overload ultimately leads to ventricular remodeling and failure. Therefore, it is unsurprising that the BK channel has arisen as a potential target for treating PH. Recently, a series of selective, synthetic BK channel agonists have proven effective in attenuating the pathophysiological progression of PH without adverse effects in animal models.
Collapse
Affiliation(s)
- Thais S Barenco-Marins
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Programa de Pós-Graduação Em Cardiologia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fernando A C Seara
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
- Instituto de Ciências Biológicas E da Saúde, Universidade Federal Rural Do Rio de Janeiro, Seropédica, RJ, Brazil.
- Programa de Pós-Graduação Multicêntrico Em Ciências Fisiológicas, Sociedade Brasileira de Fisiologia, São Paulo, Brazil.
| | - Cristiano G Ponte
- Instituto Federal de Educação, Ciências e Tecnologia do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Jose H M Nascimento
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Programa de Pós-Graduação Em Cardiologia, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
2
|
Sun WT, Xue HM, Hou HT, Chen HX, Wang J, He GW, Yang Q. Homocysteine alters vasoreactivity of human internal mammary artery by affecting the K Ca channel family. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:625. [PMID: 33987323 PMCID: PMC8106027 DOI: 10.21037/atm-20-6821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Background Hyperhomocysteinemia is an independent risk factor for atherosclerotic heart disease. We previously demonstrated that disruption of calcium-activated potassium (KCa) channel activity is involved in homocysteine-induced dilatory dysfunction of porcine coronary arteries. Recently we reported that the KCa channel family, including large-, intermediate-, and small-conductance KCa (BKCa, IKCa, and SKCa) subtypes, are abundantly expressed in human internal mammary artery (IMA). In this study, we further investigated whether homocysteine affects the expression and functionality of the KCa channel family in this commonly used graft for coronary artery bypass surgery (CABG). Methods Residual IMA segments obtained from patients undergoing CABG were studied in a myograph for the role of KCa subtypes in both vasorelaxation and vasoconstriction. The expression and distribution of KCa subtypes were detected by Western blot and immunohistochemistry. Results Both the BKCa channel activator NS1619 and the IKCa/SKCa channel activator NS309 evoked significant IMA relaxation. Homocysteine exposure suppressed NS1619-induced relaxation whereas showed no influence on NS309-induced response. Blockade of BKCa but not IKCa and SKCa subtypes significantly suppressed acetylcholine-induced relaxation and enhanced U46619-induced contraction. Homocysteine compromised the vasodilating activity of the BKCa subtype in IMA, associated with a lowered protein level of the BKCa β1-subunit. Homocysteine potentiated the role of IKCa and SKCa subtypes in mediating endothelium-dependent relaxation without affecting the expression of these channels. Conclusions Homocysteine reduces the expression of BKCa β1-subunit and compromises the vasodilating activity of BKCa channels in IMA. Unlike BKCa, IKCa and SKCa subtypes are unessential for IMA vasoregulation, whereas the loss of BKCa functionality in hyperhomocysteinemia enhances the role of IKCa and SKCa subtypes in mediating endothelial dilator function. Targeting BKCa channels may form a strategy to improve the postoperative graft performance in CABG patients with hyperhomocysteinemia who receive IMA grafting.
Collapse
Affiliation(s)
- Wen-Tao Sun
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,School of Medicine, Nankai University, Tianjin, China
| | - Hong-Mei Xue
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,School of Medicine, Nankai University, Tianjin, China.,Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Hai-Tao Hou
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Huan-Xin Chen
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jun Wang
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Guo-Wei He
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.,School of Pharmacy, Wannan Medical College, Wuhu, China.,Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA
| | - Qin Yang
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| |
Collapse
|
3
|
Glucose fluctuations promote vascular BK channels dysfunction via PKCα/NF-κB/MuRF1 signaling. J Mol Cell Cardiol 2020; 145:14-24. [PMID: 32511969 DOI: 10.1016/j.yjmcc.2020.05.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 05/27/2020] [Accepted: 05/31/2020] [Indexed: 01/01/2023]
Abstract
Glucose fluctuations may contribute to large conductance calcium activated potassium (BK) channel dysfunction. However, the underlying mechanisms remain elusive. The aim of this study was to investigate the molecular mechanisms involved in BK channel dysfunction as a result of glucose fluctuations. A rat diabetic model was established through the injection of streptozotocin. Glucose fluctuations in diabetic rats were induced via consumption and starvation. Rat coronary arteries were isolated and coronary vascular tensions were measured after three weeks. Rat coronary artery smooth muscle cells were isolated and whole-cell BK channel currents were recorded using a patch clamp technique. Human coronary artery smooth muscle cells in vitro were used to explore the underlying mechanisms. After incubation with iberiotoxin (IBTX), the Δ tensions (% Max) of rat coronary arteries in the controlled diabetes mellitus (C-DM), the uncontrolled DM (U-DM) and the DM with glucose fluctuation (GF-DM) groups were found to be 84.46 ± 5.75, 61.89 ± 10.20 and 14.77 ± 5.90, respectively (P < .05), while the current densities of the BK channels in the three groups were 43.09 ± 4.35 pA/pF, 34.23 ± 6.07 pA/pF and 17.87 ± 4.33 pA/pF, respectively (P < .05). The Δ tensions (% Max) of rat coronary arteries after applying IBTX in the GF-DM rats injected with 0.9% sodium chloride (NaCl) (GF-DM + NaCl) and the GF-DM rats injected with N-acetyl-L-cysteine (NAC) (GF-DM + NAC) groups were found to be 8.86 ± 1.09 and 48.90 ± 10.85, respectively (P < .05). Excessive oxidative stress and the activation of protein kinase C (PKC) α and nuclear factor (NF)-κB induced by glucose fluctuations promoted the decrease of BK-β1 expression, while the inhibition of reactive oxygen species (ROS), PKCα, NF-κB and muscle ring finger protein 1 (MuRF1) reversed this effect. Glucose fluctuations aggravate BK channel dysfunction via the ROS overproduction and the PKCα/NF-κB/MuRF1 signaling pathway.
Collapse
|
4
|
Man X, Liu T, Jiang Y, Zhang Z, Zhu Y, Li Z, Kong C, He J. Silencing of CARMA3 inhibits bladder cancer cell migration and invasion via deactivating β-catenin signaling pathway. Onco Targets Ther 2019; 12:6309-6322. [PMID: 31496734 PMCID: PMC6693424 DOI: 10.2147/ott.s191502] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 07/02/2019] [Indexed: 02/03/2023] Open
Abstract
Background Bladder cancer (BC) is the ninth most common cancer and the fourteenth leading death worldwide. CARD-containing MAGUK 3 (CARMA3) protein is a novel scaffold protein known to activate NF-κB pathway and is overexpressed in BC tissues. Purpose The objective of this study was to identify how CARMA3 affects the metastasis of BC cells via the β-catenin signaling pathway. Materials and methods In the present study, 5637 and T24 BC cells with stable low expression of CARMA3 were established, and their migratory and invasive capabilities were further evaluated by wound-healing and transwell assay. The activity and expression of β-catenin were determined by Luciferase assay and immunofluoresence staining. The mRNA and protein expression levels of CARMA3, matrix metallopeptidase (MMP) 9 and MMP2 were detected by quantitative real-time PCR (qRT-PCR) and Western blot analysis. The nude mouse tumor xenograft model was established for in vivo study. Results By comparison to the control cells, CARMA3-silenced cells acquired a less aggressive phenotype: decreased migration and invasion. More importantly, we confirmed that CARM3 knockdown could inhibit β-catenin mRNA and protein expression and activity, and reduce the expression and/or activity of matrix metallopeptidase (MMP) 9, MMP2 and C-myc. Also, CARM3 silencing increased E-cadherin expression and attenuated the expression of β-catenin. Moreover, we demonstrated that β-catenin overexpression reversed the inhibiting effect of CARMA3 silencing on cell invasion and migration. Furthermore, our study illustrated that knockdown of CARMA3 suppressed BC cells xenograft tumor growth in nude mice. Conclusion We demonstrated that CARMA3 contributes to the malignant phenotype of BC cells at least by activating β-catenin signaling pathway, and it may serve as a therapeutic target for clinic treatment in BC.
Collapse
Affiliation(s)
- Xiaojun Man
- Department of Urology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Institute of Urology, Department of Urology, China Medical University, Shenyang 110001, People's Republic of China
| | - Tao Liu
- Department of Urology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Institute of Urology, Department of Urology, China Medical University, Shenyang 110001, People's Republic of China
| | - Yuanjun Jiang
- Department of Urology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Institute of Urology, Department of Urology, China Medical University, Shenyang 110001, People's Republic of China
| | - Zhe Zhang
- Department of Urology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Institute of Urology, Department of Urology, China Medical University, Shenyang 110001, People's Republic of China
| | - Yuyan Zhu
- Department of Urology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Institute of Urology, Department of Urology, China Medical University, Shenyang 110001, People's Republic of China
| | - Zhenhua Li
- Department of Urology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Institute of Urology, Department of Urology, China Medical University, Shenyang 110001, People's Republic of China
| | - Chuize Kong
- Department of Urology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.,Institute of Urology, Department of Urology, China Medical University, Shenyang 110001, People's Republic of China
| | - Jiani He
- Breast Division, Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| |
Collapse
|
5
|
Balderas E, Torres NS, Rosa-Garrido M, Chaudhuri D, Toro L, Stefani E, Olcese R. MitoBK Ca channel is functionally associated with its regulatory β1 subunit in cardiac mitochondria. J Physiol 2019; 597:3817-3832. [PMID: 31173379 DOI: 10.1113/jp277769] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/03/2019] [Indexed: 01/12/2023] Open
Abstract
KEY POINTS Association of plasma membrane BKCa channels with BK-β subunits shapes their biophysical properties and physiological roles; however, functional modulation of the mitochondrial BKCa channel (mitoBKCa ) by BK-β subunits is not established. MitoBKCa -α and the regulatory BK-β1 subunit associate in mouse cardiac mitochondria. A large fraction of mitoBKCa display properties similar to that of plasma membrane BKCa when associated with BK-β1 (left-shifted voltage dependence of activation, V1/2 = -55 mV, 12 µm matrix Ca2+ ). In BK-β1 knockout mice, cardiac mitoBKCa displayed a low Po and a depolarized V1/2 of activation (+47 mV at 12 µm matrix Ca2+ ) Co-expression of BKCa with the BK-β1 subunit in HeLa cells doubled the density of BKCa in mitochondria. The present study supports the view that the cardiac mitoBKCa channel is functionally modulated by the BK-β1 subunit; proper targeting and activation of mitoBKCa shapes mitochondrial Ca2+ handling. ABSTRACT Association of the plasma membrane BKCa channel with auxiliary BK-β1-4 subunits profoundly affects the regulatory mechanisms and physiological processes in which this channel participates. However, functional association of mitochondrial BK (mitoBKCa ) with regulatory subunits is unknown. We report that mitoBKCa functionally associates with its regulatory subunit BK-β1 in adult rodent cardiomyocytes. Cardiac mitoBKCa is a calcium- and voltage-activated channel that is sensitive to paxilline with a large conductance for K+ of 300 pS. Additionally, mitoBKCa displays a high open probability (Po ) and voltage half-activation (V1/2 = -55 mV, n = 7) resembling that of plasma membrane BKCa when associated with its regulatory BK-β1 subunit. Immunochemistry assays demonstrated an interaction between mitochondrial BKCa -α and its BK-β1 subunit. Mitochondria from the BK-β1 knockout (KO) mice showed sparse mitoBKCa currents (five patches with mitoBKCa activity out of 28 total patches from n = 5 different hearts), displaying a depolarized V1/2 of activation (+47 mV in 12 µm matrix Ca2+ ). The reduced activity of mitoBKCa was accompanied by a high expression of BKCa transcript in the BK-β1 KO, suggesting a lower abundance of mitoBKCa channels in this genotype. Accordingly, BK-β1subunit increased the localization of BKDEC (i.e. the splice variant of BKCa that specifically targets mitochondria) into mitochondria by two-fold. Importantly, both paxilline-treated and BK-β1 KO mitochondria displayed a more rapid Ca2+ overload, featuring an early opening of the mitochondrial transition pore. We provide strong evidence that mitoBKCa associates with its regulatory BK-β1 subunit in cardiac mitochondria, ensuring proper targeting and activation of the mitoBKCa channel that helps to maintain mitochondrial Ca2+ homeostasis.
Collapse
Affiliation(s)
- Enrique Balderas
- Nora Eccles Harrison Cardiovascular Research & Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Natalia S Torres
- Nora Eccles Harrison Cardiovascular Research & Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Manuel Rosa-Garrido
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine.,Department of Physiology
| | - Dipayan Chaudhuri
- Nora Eccles Harrison Cardiovascular Research & Training Institute, University of Utah, Salt Lake City, UT, USA
| | - Ligia Toro
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine.,Cardiovascular Research Laboratories.,Department of Molecular and Medical Pharmacology.,Brain Research Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Enrico Stefani
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine.,Department of Physiology.,Cardiovascular Research Laboratories.,Brain Research Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Riccardo Olcese
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular Medicine.,Department of Physiology.,Cardiovascular Research Laboratories.,Brain Research Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
6
|
Nagendraprabhu P, Khatiwada S, Chaulagain S, Delhon G, Rock DL. A parapoxviral virion protein targets the retinoblastoma protein to inhibit NF-κB signaling. PLoS Pathog 2017; 13:e1006779. [PMID: 29244863 PMCID: PMC5747488 DOI: 10.1371/journal.ppat.1006779] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 12/29/2017] [Accepted: 11/28/2017] [Indexed: 12/14/2022] Open
Abstract
Poxviruses have evolved multiple strategies to subvert signaling by Nuclear Factor κB (NF-κB), a crucial regulator of host innate immune responses. Here, we describe an orf virus (ORFV) virion-associated protein, ORFV119, which inhibits NF-κB signaling very early in infection (≤ 30 min post infection). ORFV119 NF-κB inhibitory activity was found unimpaired upon translation inhibition, suggesting that virion ORFV119 alone is responsible for early interference in signaling. A C-terminal LxCxE motif in ORFV119 enabled the protein to interact with the retinoblastoma protein (pRb) a multifunctional protein best known for its tumor suppressor activity. Notably, experiments using a recombinant virus containing an ORFV119 mutation which abrogates its interaction with pRb together with experiments performed in cells lacking or with reduced pRb levels indicate that ORFV119 mediated inhibition of NF-κB signaling is largely pRb dependent. ORFV119 was shown to inhibit IKK complex activation early in infection. Consistent with IKK inhibition, ORFV119 also interacted with TNF receptor associated factor 2 (TRAF2), an adaptor protein recruited to signaling complexes upstream of IKK in infected cells. ORFV119-TRAF2 interaction was enhanced in the presence of pRb, suggesting that ORFV119-pRb complex is required for efficient interaction with TRAF2. Additionally, transient expression of ORFV119 in uninfected cells was sufficient to inhibit TNFα-induced IKK activation and NF-κB signaling, indicating that no other viral proteins are required for the effect. Infection of sheep with ORFV lacking the ORFV119 gene led to attenuated disease phenotype, indicating that ORFV119 contributes to virulence in the natural host. ORFV119 represents the first poxviral protein to interfere with NF-κB signaling through interaction with pRb.
Collapse
Affiliation(s)
- Ponnuraj Nagendraprabhu
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana- Champaign, Urbana, IL, United States of America
| | - Sushil Khatiwada
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana- Champaign, Urbana, IL, United States of America
| | - Sabal Chaulagain
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana- Champaign, Urbana, IL, United States of America
| | - Gustavo Delhon
- School of Veterinary and Biomedical Sciences, Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- * E-mail: (GD); (DLR)
| | - Daniel L. Rock
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana- Champaign, Urbana, IL, United States of America
- * E-mail: (GD); (DLR)
| |
Collapse
|
7
|
Li Y, Wang XL, Sun X, Chai Q, Li J, Thompson B, Shen WK, Lu T, Lee HC. Regulation of vascular large-conductance calcium-activated potassium channels by Nrf2 signalling. Diab Vasc Dis Res 2017; 14:353-362. [PMID: 28429615 DOI: 10.1177/1479164117703903] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BK channels are major ionic determinants of vasodilation. BK channel function is impaired in diabetic vessels due to accelerated proteolysis of its beta-1 (BK-β1) subunits in response to increased oxidative stress. The nuclear factor E2-related factor-2 (Nrf2) signalling pathway has emerged as a master regulator of cellular redox status, and we hypothesized that it plays a central role in regulating BK channel function in diabetic vessels. We found that Nrf2 expression was markedly reduced in db/db diabetic mouse aortas, and this was associated with significant downregulation of BK-β1. In addition, the muscle ring finger protein 1 (MuRF1), a known E-3 ligase targeting BK-β1 ubiquitination and proteasomal degradation, was significantly augmented. These findings were reproduced by knockdown of Nrf2 by siRNA in cultured human coronary artery smooth muscle cells. In contrast, adenoviral transfer of Nrf2 gene in these cells downregulated MuRF1 and upregulated BK-β1 expression. Activation of Nrf2 by dimethyl fumarate preserved BK-β1 expression and protected BK channel and vascular function in db/db coronary arteries. These results indicate that expression of BK-β1 is closely regulated by Nrf2 and vascular BK channel function can be restored by Nrf2 activation. Nrf2 should be considered a novel therapeutic target in the treatment of diabetic vasculopathy.
Collapse
Affiliation(s)
- Yong Li
- 1 Department of Cardiology, Affiliated Wujin Hospital of Jiangsu University, Changzhou, China
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Xiao-Li Wang
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Xiaojing Sun
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Qiang Chai
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- 3 Department of Physiology, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, P.R. China
| | - Jingchao Li
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- 4 Department of Emergency Medicine, Henan Provincial People's Hospital, Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Benjamin Thompson
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Win-Kuang Shen
- 5 Department of Cardiovascular Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Tong Lu
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Hon-Chi Lee
- 2 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
8
|
Zhang J, Li M, Zhang Z, Zhu R, Olcese R, Stefani E, Toro L. The mitochondrial BK Ca channel cardiac interactome reveals BK Ca association with the mitochondrial import receptor subunit Tom22, and the adenine nucleotide translocator. Mitochondrion 2016; 33:84-101. [PMID: 27592226 DOI: 10.1016/j.mito.2016.08.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/29/2016] [Accepted: 08/30/2016] [Indexed: 10/21/2022]
Abstract
Mitochondrial BKCa channel, mitoBKCa, regulates mitochondria function in the heart but information on its protein partnerships in cardiac mitochondria is missing. A directed proteomic approach discovered the novel interaction of BKCa with Tom22, a component of the mitochondrion outer membrane import system, and the adenine nucleotide translocator (ANT). The expressed protein partners co-immunoprecipitated and co-segregated into mitochondrial fractions in HEK293T cells. The BKCa 50 amino acid splice insert, DEC, facilitated BKCa interaction with ANT. Further, BKCa transmembrane domain was required for the association with both Tom22 and ANT. The results serve as a working framework to understand mitoBKCa import and functional relationships.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Anesthesiology, Division of Molecular Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Min Li
- Department of Anesthesiology, Division of Molecular Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zhu Zhang
- Department of Anesthesiology, Division of Molecular Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ronghui Zhu
- Department of Anesthesiology, Division of Molecular Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Riccardo Olcese
- Department of Anesthesiology, Division of Molecular Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Physiology, University of California, Los Angeles, Los Angeles, CA, USA; Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA; Cardiovascular Research Laboratory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Enrico Stefani
- Department of Anesthesiology, Division of Molecular Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Physiology, University of California, Los Angeles, Los Angeles, CA, USA; Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA; Cardiovascular Research Laboratory, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ligia Toro
- Department of Anesthesiology, Division of Molecular Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Molecular & Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA; Brain Research Institute, University of California, Los Angeles, Los Angeles, CA, USA; Cardiovascular Research Laboratory, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Chen Q, Tao J, Hei H, Li F, Wang Y, Peng W, Zhang X. Up-Regulatory Effects of Curcumin on Large Conductance Ca2+-Activated K+ Channels. PLoS One 2015; 10:e0144800. [PMID: 26672753 PMCID: PMC4682634 DOI: 10.1371/journal.pone.0144800] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 11/24/2015] [Indexed: 12/21/2022] Open
Abstract
Large conductance Ca2+-activated potassium channels (BK) are targets for research that explores therapeutic means to various diseases, owing to the roles of the channels in mediating multiple physiological processes in various cells and tissues. We investigated the pharmacological effects of curcumin, a compound isolated from the herb Curcuma longa, on BK channels. As recorded by whole-cell patch-clamp, curcumin increased BK (α) and BK (α+β1) currents in transfected HEK293 cells as well as the current density of BK in A7r5 smooth muscle cells in a dose-dependent manner. By incubating with curcumin for 24 hours, the current density of exogenous BK (α) in HEK293 cells and the endogenous BK in A7r5 cells were both enhanced notably, though the steady-state activation of the channels did not shift significantly, except for BK (α+β1). Curcumin up-regulated the BK protein expression without changing its mRNA level in A7r5 cells. The surface expression and the half-life of BK channels were also increased by curcumin in HEK293 cells. These effects of curcumin were abolished by MG-132, a proteasome inhibitor. Curcumin also increased ERK 1/2 phosphorylation, while inhibiting ERK by U0126 attenuated the curcumin-induced up-regulation of BK protein expression. We also observed that the curcumin-induced relaxation in the isolated rat aortic rings was significantly attenuated by paxilline, a BK channel specific blocker. These results show that curcumin enhances the activity of the BK channels by interacting with BK directly as well as enhancing BK protein expression through inhibiting proteasomal degradation and activating ERK signaling pathway. The findings suggest that curcumin is a potential BK channel activator and provide novel insight into its complicated pharmacological effects and the underlying mechanisms.
Collapse
Affiliation(s)
- Qijing Chen
- Department of Pharmacology, School of Pharmacy, Fudan University, 826 Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Jie Tao
- Department of Pharmacology, School of Pharmacy, Fudan University, 826 Zhangheng Road, Pudong New District, Shanghai, 201203, China
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine,164 Lanxi road, Shanghai, 200062, China
| | - Hongya Hei
- Department of Pharmacology, School of Pharmacy, Fudan University, 826 Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Fangping Li
- Department of Pharmacology, School of Pharmacy, Fudan University, 826 Zhangheng Road, Pudong New District, Shanghai, 201203, China
| | - Yunman Wang
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine,164 Lanxi road, Shanghai, 200062, China
| | - Wen Peng
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine,164 Lanxi road, Shanghai, 200062, China
- * E-mail: (XZ); (WP)
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, 826 Zhangheng Road, Pudong New District, Shanghai, 201203, China
- * E-mail: (XZ); (WP)
| |
Collapse
|
10
|
Clements RT, Terentyev D, Sellke FW. Ca 2+-Activated K + Channels as Therapeutic Targets for Myocardial and Vascular Protection. Circ J 2015; 79:455-62. [DOI: 10.1253/circj.cj-15-0015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Richard T. Clements
- Department of Surgery, Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University
| | - Dmitry Terentyev
- Department of Medicine, Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University
| | - Frank W. Sellke
- Department of Surgery, Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University
| |
Collapse
|
11
|
Toro L, Li M, Zhang Z, Singh H, Wu Y, Stefani E. MaxiK channel and cell signalling. Pflugers Arch 2014; 466:875-86. [PMID: 24077696 DOI: 10.1007/s00424-013-1359-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 09/09/2013] [Accepted: 09/10/2013] [Indexed: 01/23/2023]
Abstract
The large-conductance Ca2+- and voltage-activated K+ (MaxiK, BK, BKCa, Slo1, KCa1.1) channel role in cell signalling is becoming apparent as we learn how the channel interacts with a multiplicity of proteins not only at the plasma membrane but also in intracellular organelles including the endoplasmic reticulum, nucleus, and mitochondria. In this review, we focus on the interactions of MaxiK channels with seven-transmembrane G protein-coupled receptors and discuss information suggesting that, the channel big C-terminus may act as the nucleus of signalling molecules including kinases relevant for cell death and survival. Increasing evidence indicates that the channel is able to associate with a variety of receptors including β-adrenergic receptors, G protein-coupled estrogen receptors, acetylcholine receptors, thromboxane A2 receptors, and angiotensin II receptors, which highlights the varied functions that the channel has (or may have) not only in regulating contraction/relaxation of muscle cells or neurotransmission in the brain but also in cell metabolism, proliferation, migration, and gene expression. In line with this view, MaxiK channels have been implicated in obesity and in brain, prostate, and mammary cancers. A better understanding on the molecular mechanisms underlying or triggered by MaxiK channel abnormalities like overexpression in certain cancers may lead to new therapeutics to prevent devastating diseases.
Collapse
|
12
|
Rottlerin-induced BKCa channel activation impairs specific contractile responses and promotes vasodilation. Ann Thorac Surg 2014; 99:626-34. [PMID: 25527424 DOI: 10.1016/j.athoracsur.2014.07.091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 07/16/2014] [Accepted: 07/21/2014] [Indexed: 12/27/2022]
Abstract
BACKGROUND Activation of large conductance calcium-activated potassium (BKCa) channels is cardioprotective for ischemic injury and can enhance vasorelaxation. Rottlerin has recently been identified as a potent BKCa activator. We demonstrated that rottlerin improves cardiac function and increases coronary flow when used as a cardioplegia additive in rat and mouse models of cardioplegic arrest and reperfusion. In this study we examined the effectiveness and specificity of the putative BKCa activator rottlerin on vascular reactivity in response to specific contractile and dilatory agonists. METHODS Aortic rings from wild-type (wt) and BKCa knock-out (KO) mice were mounted in a tissue bath with force transducers. The vasodilatory effect of rottlerin was evaluated after pre-constriction with U46619. Dose responses to the contractile agonists U46619 and phenylephrine (PE), and vasodilation responses to rottlerin, hydrogen sulfide (H2S), and sodium nitroprusside (SNP) were performed after pretreatment with rottlerin. Similar studies were performed in pig coronary vessels. RESULTS The BKCa KO mouse aortic rings exhibited spontaneous contraction and had greater contractile responses to U46619 and reduced vasodilation to SNP compared with wt mice. The wt and KO responses to phenylephrine were similar. Rottlerin dose dependently dilated wild-type vessels, but not in BKCa KO animals. Pretreatment with rottlerin caused depressed U46619 responses, but had no effect on PE, SNP, or H2S-mediated responses. However, pig coronary vessels pretreated with rottlerin exhibited reduced contractile responses and enhanced nitric oxide-dependent dilation. CONCLUSIONS Rottlerin directly causes vasodilation through BKCa channel dependent mechanisms. The BKCa channel activator pretreatment enhances vasodilatory responses and impairs specific vasoconstrictive agonists.
Collapse
|
13
|
Dopico AM, Bukiya AN. Lipid regulation of BK channel function. Front Physiol 2014; 5:312. [PMID: 25202277 PMCID: PMC4141547 DOI: 10.3389/fphys.2014.00312] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/31/2014] [Indexed: 01/11/2023] Open
Abstract
This mini-review focuses on lipid modulation of BK (MaxiK, BKCa) current by a direct interaction between lipid and the BK subunits and/or their immediate lipid environment. Direct lipid-BK protein interactions have been proposed for fatty and epoxyeicosatrienoic acids, phosphoinositides and cholesterol, evidence for such action being less clear for other lipids. BK α (slo1) subunits are sufficient to support current perturbation by fatty and epoxyeicosatrienoic acids, glycerophospholipids and cholesterol, while distinct BK β subunits seem necessary for current modulation by most steroids. Subunit domains or amino acids that participate in lipid action have been identified in a few cases: hslo1 Y318, cerebral artery smooth muscle (cbv1) R334,K335,K336, cbv1 seven cytosolic CRAC domains, slo1 STREX and β1 T169,L172,L173 for docosahexaenoic acid, PIP2, cholesterol, sulfatides, and cholane steroids, respectively. Whether these protein motifs directly bind lipids or rather transmit the energy of lipid binding to other areas and trigger protein conformation change remains unresolved. The impact of direct lipid-BK interaction on physiology is briefly discussed.
Collapse
Affiliation(s)
- Alex M Dopico
- Department of Pharmacology, The University of Tennessee Health Science Center Memphis, TN, USA
| | - Anna N Bukiya
- Department of Pharmacology, The University of Tennessee Health Science Center Memphis, TN, USA
| |
Collapse
|
14
|
Zhang Z, Li M, Lu R, Alioua A, Stefani E, Toro L. The angiotensin II type 1 receptor (AT1R) closely interacts with large conductance voltage- and Ca2+-activated K+ (BK) channels and inhibits their activity independent of G-protein activation. J Biol Chem 2014; 289:25678-89. [PMID: 25070892 DOI: 10.1074/jbc.m114.595603] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Angiotensin II (ANG-II) and BK channels play important roles in the regulation of blood pressure. In arterial smooth muscle, ANG-II inhibits BK channels, but the underlying molecular mechanisms are unknown. Here, we first investigated whether ANG-II utilizes its type 1 receptor (AT1R) to modulate BK activity. Pharmacological, biochemical, and molecular evidence supports a role for AT1R. In renal arterial myocytes, the AT1R antagonist losartan (10 μM) abolished the ANG-II (1 μM)-induced reduction of whole cell BK currents, and BK channels and ANG-II receptors were found to co-localize at the cell periphery. We also found that BK inhibition via ANG-II-activated AT1R was independent of G-protein activation (assessed with 500 μM GDPβS). In BK-expressing HEK293T cells, ANG-II (1 μM) also induced a reduction of BK currents, which was contingent on AT1R expression. The molecular mechanisms of AT1R and BK channel coupling were investigated in co-transfected cells. Co-immunoprecipitation showed formation of a macromolecular complex, and live immunolabeling demonstrated that both proteins co-localized at the plasma membrane with high proximity indexes as in arterial myocytes. Consistent with a close association, we discovered that the sole AT1R expression could decrease BK channel voltage sensitivity. Truncated BK proteins revealed that the voltage-sensing conduction cassette is sufficient for BK-AT1R association. Finally, C-terminal yellow and cyan fluorescent fusion proteins, AT1R-YFP and BK-CFP, displayed robust co-localized Förster resonance energy transfer, demonstrating intermolecular interactions at their C termini. Overall, our results strongly suggest that AT1R regulates BK channels through a close protein-protein interaction involving multiple BK regions and independent of G-protein activation.
Collapse
Affiliation(s)
- Zhu Zhang
- From the Departments of Anesthesiology
| | - Min Li
- From the Departments of Anesthesiology
| | - Rong Lu
- From the Departments of Anesthesiology
| | | | - Enrico Stefani
- From the Departments of Anesthesiology, Physiology, the Brain Research Institute, and the Cardiovascular Research Laboratory, University of California, Los Angeles, California 90095
| | - Ligia Toro
- From the Departments of Anesthesiology, the Brain Research Institute, and the Cardiovascular Research Laboratory, University of California, Los Angeles, California 90095 Molecular and Medical Pharmacology, and
| |
Collapse
|
15
|
Yi F, Wang H, Chai Q, Wang X, Shen WK, Willis MS, Lee HC, Lu T. Regulation of large conductance Ca2+-activated K+ (BK) channel β1 subunit expression by muscle RING finger protein 1 in diabetic vessels. J Biol Chem 2014; 289:10853-10864. [PMID: 24570002 DOI: 10.1074/jbc.m113.520940] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The large conductance Ca(2+)-activated K(+) (BK) channel, expressed abundantly in vascular smooth muscle cells (SMCs), is a key determinant of vascular tone. BK channel activity is tightly regulated by its accessory β1 subunit (BK-β1). However, BK channel function is impaired in diabetic vessels by increased ubiquitin/proteasome-dependent BK-β1 protein degradation. Muscle RING finger protein 1 (MuRF1), a muscle-specific ubiquitin ligase, is implicated in many cardiac and skeletal muscle diseases. However, the role of MuRF1 in the regulation of vascular BK channel and coronary function has not been examined. In this study, we hypothesized that MuRF1 participated in BK-β1 proteolysis, leading to the down-regulation of BK channel activation and impaired coronary function in diabetes. Combining patch clamp and molecular biological approaches, we found that MuRF1 expression was enhanced, accompanied by reduced BK-β1 expression, in high glucose-cultured human coronary SMCs and in diabetic vessels. Knockdown of MuRF1 by siRNA in cultured human SMCs attenuated BK-β1 ubiquitination and increased BK-β1 expression, whereas adenoviral expression of MuRF1 in mouse coronary arteries reduced BK-β1 expression and diminished BK channel-mediated vasodilation. Physical interaction between the N terminus of BK-β1 and the coiled-coil domain of MuRF1 was demonstrated by pulldown assay. Moreover, MuRF1 expression was regulated by NF-κB. Most importantly, pharmacological inhibition of proteasome and NF-κB activities preserved BK-β1 expression and BK-channel-mediated coronary vasodilation in diabetic mice. Hence, our results provide the first evidence that the up-regulation of NF-κB-dependent MuRF1 expression is a novel mechanism that leads to BK channelopathy and vasculopathy in diabetes.
Collapse
Affiliation(s)
- Fu Yi
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xian 710032, China
| | - Huan Wang
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Qiang Chai
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Xiaoli Wang
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Win-Kuang Shen
- Department of Internal Medicine, Mayo Clinic, Scottsdale, Arizona 85259
| | - Monte S Willis
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Hon-Chi Lee
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | - Tong Lu
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota 55905.
| |
Collapse
|
16
|
Ellinsworth DC, Shukla N, Fleming I, Jeremy JY. Interactions between thromboxane A₂, thromboxane/prostaglandin (TP) receptors, and endothelium-derived hyperpolarization. Cardiovasc Res 2014; 102:9-16. [PMID: 24469536 DOI: 10.1093/cvr/cvu015] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Endothelium-dependent smooth muscle hyperpolarization (EDH) increasingly predominates over endothelium-derived nitric oxide (NO) as a participant in vasodilation as vessel size decreases. Its underlying nature is highly variable between vessel types, species, disease states, and exact experimental conditions, and is variably mediated by one or more transferable endothelium-derived hyperpolarizing factors and/or the electrotonic spread of endothelial hyperpolarization into the media via gap junctions. Although generally regarded (and studied) as a mechanism that is independent of NO and prostanoids, evidence has emerged that the endothelium-derived contracting factor and prostanoid thromboxane A2 can modulate several signalling components central to EDH, and therefore potentially curtail vasodilation through mechanisms that are distinct from those putatively involved in direct smooth muscle contraction. Notably, vascular production of thromboxane A2 is elevated in a number of cardiovascular disease states that promote endothelial dysfunction. This review will therefore discuss the mechanisms through which thromboxane A2 interacts with and modulates EDH, and will also consider the implications of such cross-talk in vasodilator control in health and disease.
Collapse
Affiliation(s)
- David C Ellinsworth
- Bristol Heart Institute, University of Bristol, Queens Building Level 7, Upper Maudlin St, Bristol Royal Infirmary, Upper Maudlin Street, Bristol BS2 8HW, UK
| | | | | | | |
Collapse
|