1
|
Agarwal S, Kim ED, Lee S, Simon A, Accardi A, Nimigean CM. Ball-and-chain inactivation of a human large conductance calcium-activated potassium channel. Nat Commun 2025; 16:1769. [PMID: 39971906 PMCID: PMC11840039 DOI: 10.1038/s41467-025-56844-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/03/2025] [Indexed: 02/21/2025] Open
Abstract
BK channels are large-conductance calcium (Ca2+)-activated potassium channels crucial for neuronal excitability, muscle contraction, and neurotransmitter release. The pore-forming (α) subunits co-assemble with auxiliary (β and γ) subunits that modulate their function. Previous studies demonstrated that the N-termini of β2-subunits can inactivate BK channels, but with no structural correlate. Here, we investigate BK β2-subunit inactivation using cryo-electron microscopy, electrophysiology and molecular dynamics simulations. We find that the β2 N-terminus occludes the pore only in the Ca2+-bound open state, via a ball-and-chain mechanism. The first three hydrophobic residues of β2 are crucial for occlusion, while the remainder of the N-terminus remains flexible. Neither the closed channel conformation obtained in the absence of Ca2+ nor an intermediate conformation found in the presence of Ca2+ show density for the N-terminus of the β2 subunit in their pore, likely due to narrower side access portals preventing their entry into the channel pore.
Collapse
Affiliation(s)
- Shubhangi Agarwal
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Elizabeth D Kim
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Sangyun Lee
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Alexander Simon
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Alessio Accardi
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA
| | - Crina M Nimigean
- Department of Anesthesiology, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA.
- Department of Physiology and Biophysics, Weill Cornell Medical College, 1300 York Ave, New York, NY, USA.
| |
Collapse
|
2
|
Ancatén-González C, Meza RC, Gonzalez-Sanabria N, Segura I, Alcaino A, Peña-Pichicoi A, Latorre R, Chiu CQ, Chávez AE. BK channels mediate a presynaptic form of mGluR-LTD in the neonatal hippocampus. Proc Natl Acad Sci U S A 2025; 122:e2411506122. [PMID: 39773031 PMCID: PMC11745352 DOI: 10.1073/pnas.2411506122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025] Open
Abstract
BK channels can control neuronal function, but their functional relevance in activity-dependent changes of synaptic function remains elusive. Here, we report that repetitive low-frequency stimulation activates BK channels through 12(S)HPETE, an arachidonic acid metabolite, produced downstream of postsynaptic metabotropic glutamate receptors (mGluRs) to trigger long-term depression (LTD) at CA3-CA1 synapses in hippocampal slices from P7-P10 mice. Activation of BK channels is subunit specific, as paxilline but not iberiotoxin blocked mGluR-LTD. Also, 12(S)HPETE does not change the electrophysiological properties of the BK channel when the BKα subunit is expressed alone but increases the channel open probability when the BKα is coexpressed with the β4-subunit. Our findings reveal an interaction between 12(S)HPETE and BK channels to regulate synaptic strength at central synapses and increase our understanding of the mechanisms underlying mGluR-LTD in the neonatal hippocampus that likely contribute to circuit maturation necessary for learning.
Collapse
Affiliation(s)
- Carlos Ancatén-González
- Programa de Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Rodrigo C. Meza
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Naileth Gonzalez-Sanabria
- Programa de Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Ignacio Segura
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Alejandro Alcaino
- Programa de Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Antonio Peña-Pichicoi
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Ramón Latorre
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Chiayu Q. Chiu
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| | - Andrés E. Chávez
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso2340000, Chile
| |
Collapse
|
3
|
Nelapudi N, Boskind M, Hu XQ, Mallari D, Chan M, Wilson D, Romero M, Albert-Minckler E, Zhang L, Blood AB, Wilson CG, Puglisi JL, Wilson SM. Long-term hypoxia modulates depolarization activation of BK Ca currents in fetal sheep middle cerebral arterial myocytes. Front Physiol 2024; 15:1479882. [PMID: 39563935 PMCID: PMC11573761 DOI: 10.3389/fphys.2024.1479882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/17/2024] [Indexed: 11/21/2024] Open
Abstract
Introduction Previous evidence indicates that gestational hypoxia disrupts cerebrovascular development, increasing the risk of intracranial hemorrhage and stroke in the newborn. Due to the role of cytosolic Ca2+ in regulating vascular smooth muscle (VSM) tone and fetal cerebrovascular blood flow, understanding Ca2+ signals can offer insight into the pathophysiological disruptions taking place in hypoxia-related perinatal cerebrovascular disease. This study aimed to determine the extent to which gestational hypoxia disrupts local Ca2+ sparks and whole-cell Ca2+ signals and coupling with BKCa channel activity. Methods Confocal imaging of cytosolic Ca2+ and recording BKCa currents of fetal sheep middle cerebral arterial (MCA) myocytes was performed. MCAs were isolated from term fetal sheep (∼140 days of gestation) from ewes held at low- (700 m) and high-altitude (3,801 m) hypoxia (LTH) for 100+ days of gestation. Arteries were depolarized with 30 mM KCl (30K), in the presence or absence of 10 μM ryanodine (Ry), to block RyR mediated Ca2+ release. Results Membrane depolarization increased Ry-sensitive Ca2+ spark frequency in normoxic and LTH groups along with BKCa activity. LTH reduced Ca2+ spark and whole-cell Ca2+ activity and induced a large leftward shift in the voltage-dependence of BKCa current activation. The influence of LTH on the spatial and temporal aspects of Ca2+ sparks and whole-cell Ca2+ responses varied. Discussion Overall, LTH attenuates Ca2+ signaling while increasing the coupling of Ca2+ sparks to BKCa activity; a process that potentially helps maintain oxygen delivery to the developing brain.
Collapse
Affiliation(s)
- Nikitha Nelapudi
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Madison Boskind
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Xiang-Qun Hu
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - David Mallari
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Michelle Chan
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Devin Wilson
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Monica Romero
- Advanced Imaging and Microscopy Core, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Eris Albert-Minckler
- Advanced Imaging and Microscopy Core, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Lubo Zhang
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Arlin B Blood
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Christopher G Wilson
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Jose Luis Puglisi
- Department of Biostatistics, California Northstate University School of Medicine, Elk Grove, CA, United States
| | - Sean M Wilson
- Lawrence D Longo Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA, United States
- Advanced Imaging and Microscopy Core, Loma Linda University School of Medicine, Loma Linda, CA, United States
| |
Collapse
|
4
|
Uchiumi O, Zou J, Yamaki S, Hori Y, Ono M, Yamamoto R, Kato N. Disruption of sphingomyelin synthase 2 gene alleviates cognitive impairment in a mouse model of Alzheimer's disease. Brain Res 2024; 1835:148934. [PMID: 38609029 DOI: 10.1016/j.brainres.2024.148934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
The membrane raft accommodates the key enzymes synthesizing amyloid β (Aβ). One of the two characteristic components of the membrane raft, cholesterol, is well known to promote the key enzymes that produce amyloid-β (Aβ) and exacerbate Alzheimer's disease (AD) pathogenesis. Given that the raft is a physicochemical platform for the sound functioning of embedded bioactive proteins, the other major lipid component sphingomyelin may also be involved in AD. Here we knocked out the sphingomyelin synthase 2 gene (SMS2) in 3xTg AD model mice by hybridization, yielding SMS2KO mice (4S mice). The novel object recognition test in 9/10-month-old 4S mice showed that cognitive impairment in 3xTg mice was alleviated by SMS2KO, though performance in the Morris water maze (MWM) was not improved. The tail suspension test detected a depressive trait in 4S mice, which may have hindered the manifestation of performance in the wet, stressful environment of MWM. In the hippocampal CA1, hyperexcitability in 3xTg was also found alleviated by SMS2KO. In the hippocampal dentate gyrus of 4S mice, the number of neurons positive with intracellular Aβ or its precursor proteins, the hallmark of young 3xTg mice, is reduced to one-third, suggesting an SMS2KO-led suppression of syntheses of those peptides in the dentate gyrus. Although we previously reported that large-conductance calcium-activated potassium (BK) channels are suppressed in 3xTg mice and their recovery relates to cognitive amelioration, no changes occurred by hybridization. Sphingomyelin in the membrane raft may serve as a novel target for AD drugs.
Collapse
Affiliation(s)
- Osamu Uchiumi
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Jingyu Zou
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan; First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Sachiko Yamaki
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Yoshie Hori
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Munenori Ono
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Ryo Yamamoto
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Nobuo Kato
- Department of Physiology, Kanazawa Medical University, Ishikawa 920-0293, Japan.
| |
Collapse
|
5
|
Olszewska AM, Zmijewski MA. Genomic and non-genomic action of vitamin D on ion channels - Targeting mitochondria. Mitochondrion 2024; 77:101891. [PMID: 38692383 DOI: 10.1016/j.mito.2024.101891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/03/2024]
Abstract
Recent studies revealed that mitochondria are not only a place of vitamin D3 metabolism but also direct or indirect targets of its activities. This review summarizes current knowledge on the regulation of ion channels from plasma and mitochondrial membranes by the active form of vitamin D3 (1,25(OH)2D3). 1,25(OH)2D3, is a naturally occurring hormone with pleiotropic activities; implicated in the modulation of cell differentiation, and proliferation and in the prevention of various diseases, including cancer. Many experimental data indicate that 1,25(OH)2D3 deficiency induces ionic remodeling and 1,25(OH)2D3 regulates the activity of multiple ion channels. There are two main theories on how 1,25(OH)2D3 can modify the function of ion channels. First, describes the involvement of genomic pathways of response to 1,25(OH)2D3 in the regulation of the expression of the genes encoding channels, their auxiliary subunits, or additional regulators. Interestingly, intracellular ion channels, like mitochondrial, are encoded by the same genes as plasma membrane channels. Therefore, the comprehensive genomic regulation of the channels from these two different cellular compartments we analyzed using a bioinformatic approach. The second theory explores non-genomic pathways of vitamin D3 activities. It was shown, that 1,25(OH)2D3 indirectly regulates enzymes that impact ion channels, change membrane physical properties, or directly bind to channel proteins. In this article, the involvement of genomic and non-genomic pathways regulated by 1,25(OH)2D3 in the modulation of the levels and activity of plasma membrane and mitochondrial ion channels was investigated by an extensive review of the literature and analysis of the transcriptomic data using bioinformatics.
Collapse
Affiliation(s)
- A M Olszewska
- Department of Histology, Medical University of Gdansk, 1a Debinki, 80-211 Gdansk, Poland
| | - M A Zmijewski
- Department of Histology, Medical University of Gdansk, 1a Debinki, 80-211 Gdansk, Poland.
| |
Collapse
|
6
|
Fan C, Flood E, Sukomon N, Agarwal S, Allen TW, Nimigean CM. Calcium-gated potassium channel blockade via membrane-facing fenestrations. Nat Chem Biol 2024; 20:52-61. [PMID: 37653172 PMCID: PMC10847966 DOI: 10.1038/s41589-023-01406-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/18/2023] [Indexed: 09/02/2023]
Abstract
Quaternary ammonium blockers were previously shown to bind in the pore to block both open and closed conformations of large-conductance calcium-activated potassium (BK and MthK) channels. Because blocker entry was assumed through the intracellular entryway (bundle crossing), closed-pore access suggested that the gate was not at the bundle crossing. Structures of closed MthK, a Methanobacterium thermoautotrophicum homolog of BK channels, revealed a tightly constricted intracellular gate, leading us to investigate the membrane-facing fenestrations as alternative pathways for blocker access directly from the membrane. Atomistic free energy simulations showed that intracellular blockers indeed access the pore through the fenestrations, and a mutant channel with narrower fenestrations displayed no closed-state TPeA block at concentrations that blocked the wild-type channel. Apo BK channels display similar fenestrations, suggesting that blockers may use them as access paths into closed channels. Thus, membrane fenestrations represent a non-canonical pathway for selective targeting of specific channel conformations, opening novel ways to selectively drug BK channels.
Collapse
Affiliation(s)
- Chen Fan
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, Sweden
| | - Emelie Flood
- School of Science, RMIT University, Melbourne, Victoria, Australia
- Schrödinger, Inc., New York, NY, USA
| | - Nattakan Sukomon
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Shubhangi Agarwal
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA
| | - Toby W Allen
- School of Science, RMIT University, Melbourne, Victoria, Australia.
| | - Crina M Nimigean
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA.
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
7
|
North KC, Shaw AA, Bukiya AN, Dopico AM. Progesterone activation of β 1-containing BK channels involves two binding sites. Nat Commun 2023; 14:7248. [PMID: 37945687 PMCID: PMC10636063 DOI: 10.1038/s41467-023-42827-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Progesterone (≥1 µM) is used in recovery of cerebral ischemia, an effect likely contributed to by cerebrovascular dilation. The targets of this progesterone action are unknown. We report that micromolar (µM) progesterone activates mouse cerebrovascular myocyte BK channels; this action is lost in β1-/- mice myocytes and in lipid bilayers containing BK α subunit homomeric channels but sustained on β1/β4-containing heteromers. Progesterone binds to both regulatory subunits, involving two steroid binding sites conserved in β1-β4: high-affinity (sub-µM), which involves Trp87 in β1 loop, and low-affinity (µM) defined by TM1 Tyr32 and TM2 Trp163. Thus progesterone, but not its oxime, bridges TM1-TM2. Mutation of the high-affinity site blunts channel activation by progesterone underscoring a permissive role of the high-affinity site: progesterone binding to this site enables steroid binding at the low-affinity site, which activates the channel. In support of our model, cerebrovascular dilation evoked by μM progesterone is lost by mutating Tyr32 or Trp163 in β1 whereas these mutations do not affect alcohol-induced cerebrovascular constriction. Furthermore, this alcohol action is effectively counteracted both in vitro and in vivo by progesterone but not by its oxime.
Collapse
Affiliation(s)
- Kelsey C North
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Andrew A Shaw
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Anna N Bukiya
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Alex M Dopico
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA.
| |
Collapse
|
8
|
Mysiewicz SC, Hawks SM, Bukiya AN, Dopico AM. Differential Functional Contribution of BK Channel Subunits to Aldosterone-Induced Channel Activation in Vascular Smooth Muscle and Eventual Cerebral Artery Dilation. Int J Mol Sci 2023; 24:ijms24108704. [PMID: 37240049 DOI: 10.3390/ijms24108704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/03/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Calcium/voltage-activated potassium channels (BK) control smooth muscle (SM) tone and cerebral artery diameter. They include channel-forming α and regulatory β1 subunits, the latter being highly expressed in SM. Both subunits participate in steroid-induced modification of BK activity: β1 provides recognition for estradiol and cholanes, resulting in BK potentiation, whereas α suffices for BK inhibition by cholesterol or pregnenolone. Aldosterone can modify cerebral artery function independently of its effects outside the brain, yet BK involvement in aldosterone's cerebrovascular action and identification of channel subunits, possibly involved in steroid action, remains uninvestigated. Using microscale thermophoresis, we demonstrated that each subunit type presents two recognition sites for aldosterone: at 0.3 and ≥10 µM for α and at 0.3-1 µM and ≥100 µM for β1. Next, we probed aldosterone on SM BK activity and diameter of middle cerebral artery (MCA) isolated from β1-/- vs. wt mice. Data showed that β1 leftward-shifted aldosterone-induced BK activation, rendering EC50~3 μM and ECMAX ≥ 10 μM, at which BK activity increased by 20%. At similar concentrations, aldosterone mildly yet significantly dilated MCA independently of circulating and endothelial factors. Lastly, aldosterone-induced MCA dilation was lost in β1-/- mice. Therefore, β1 enables BK activation and MCA dilation by low µM aldosterone.
Collapse
Affiliation(s)
- Steven C Mysiewicz
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Sydney M Hawks
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Anna N Bukiya
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Alex M Dopico
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| |
Collapse
|
9
|
Mysiewicz S, North KC, Moreira L, Odum SJ, Bukiya AN, Dopico AM. Interspecies and regional variability of alcohol action on large cerebral arteries: regulation by KCNMB1 proteins. Am J Physiol Regul Integr Comp Physiol 2023; 324:R480-R496. [PMID: 36717168 PMCID: PMC10027090 DOI: 10.1152/ajpregu.00103.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 02/01/2023]
Abstract
Alcohol intake leading to blood ethanol concentrations (BEC) ≥ legal intoxication modifies brain blood flow with increases in some regions and decreases in others. Brain regions receive blood from the Willis' circle branches: anterior, middle (MCA) and posterior cerebral (PCA), and basilar (BA) arteries. Rats and mice have been used to identify the targets mediating ethanol-induced effects on cerebral arteries, with conclusions being freely interchanged, albeit data were obtained in different species/arterial branches. We tested whether ethanol action on cerebral arteries differed between male rat and mouse and/or across different brain regions and identified the targets of alcohol action. In both species and all Willis' circle branches, ethanol evoked reversible and concentration-dependent constriction (EC50s ≈ 37-86 mM; below lethal BEC in alcohol-naïve humans). Although showing similar constriction to depolarization, both species displayed differential responses to ethanol: in mice, MCA constriction was highly sensitive to the presence/absence of the endothelium, whereas in rat PCA was significantly more sensitive to ethanol than its mouse counterpart. In the rat, but not the mouse, BA was more ethanol sensitive than other branches. Both interspecies and regional variability were ameliorated by endothelium. Selective large conductance (BK) channel block in de-endothelialized vessels demonstrated that these channels were the effectors of alcohol-induced cerebral artery constriction across regions and species. Variabilities in alcohol actions did not fully matched KCNMB1 expression across vessels. However, immunofluorescence data from KCNMB1-/- mouse arteries electroporated with KCNMB1-coding cDNA demonstrate that KCNMB1 proteins, which regulate smooth muscle (SM) BK channel function and vasodilation, regulate interspecies and regional variability of brain artery responses to alcohol.
Collapse
Affiliation(s)
- Steven Mysiewicz
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Kelsey C North
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Luiz Moreira
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Schyler J Odum
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Anna N Bukiya
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Alex M Dopico
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| |
Collapse
|
10
|
Potassium channelopathies associated with epilepsy-related syndromes and directions for therapeutic intervention. Biochem Pharmacol 2023; 208:115413. [PMID: 36646291 DOI: 10.1016/j.bcp.2023.115413] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
A number of mutations to members of several CNS potassium (K) channel families have been identified which result in rare forms of neonatal onset epilepsy, or syndromes of which one prominent characteristic is a form of epilepsy. Benign Familial Neonatal Convulsions or Seizures (BFNC or BFNS), also referred to as Self-Limited Familial Neonatal Epilepsy (SeLNE), results from mutations in 2 members of the KV7 family (KCNQ) of K channels; while generally self-resolving by about 15 weeks of age, these mutations significantly increase the probability of generalized seizure disorders in the adult, in some cases they result in more severe developmental syndromes. Epilepsy of Infancy with Migrating Focal Seizures (EIMSF), or Migrating Partial Seizures of Infancy (MMPSI), is a rare severe form of epilepsy linked primarily to gain of function mutations in a member of the sodium-dependent K channel family, KCNT1 or SLACK. Finally, KCNMA1 channelopathies, including Liang-Wang syndrome (LIWAS), are rare combinations of neurological symptoms including seizure, movement abnormalities, delayed development and intellectual disabilities, with Liang-Wang syndrome an extremely serious polymalformative syndrome with a number of neurological sequelae including epilepsy. These are caused by mutations in the pore-forming subunit of the large-conductance calcium-activated K channel (BK channel) KCNMA1. The identification of these rare but significant channelopathies has resulted in a resurgence of interest in their treatment by direct pharmacological or genetic modulation. We will briefly review the genetics, biophysics and pharmacology of these K channels, their linkage with the 3 syndromes described above, and efforts to more effectively target these syndromes.
Collapse
|
11
|
Vaithianathan T, Schneider EH, Bukiya AN, Dopico AM. Cholesterol and PIP 2 Modulation of BK Ca Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:217-243. [PMID: 36988883 PMCID: PMC10683925 DOI: 10.1007/978-3-031-21547-6_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Ca2+/voltage-gated, large conductance K+ channels (BKCa) are formed by homotetrameric association of α (slo1) subunits. Their activity, however, is suited to tissue-specific physiology largely due to their association with regulatory subunits (β and γ types), chaperone proteins, localized signaling, and the channel's lipid microenvironment. PIP2 and cholesterol can modulate BKCa activity independently of downstream signaling, yet activating Ca2+i levels and regulatory subunits control ligand action. At physiological Ca2+i and voltages, cholesterol and PIP2 reduce and increase slo1 channel activity, respectively. Moreover, slo1 proteins provide sites that seem to recognize cholesterol and PIP2: seven CRAC motifs in the slo1 cytosolic tail and a string of positively charged residues (Arg329, Lys330, Lys331) immediately after S6, respectively. A model that could explain the modulation of BKCa activity by cholesterol and/or PIP2 is hypothesized. The roles of additional sites, whether in slo1 or BKCa regulatory subunits, for PIP2 and/or cholesterol to modulate BKCa function are also discussed.
Collapse
Affiliation(s)
- Thirumalini Vaithianathan
- Department Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Elizabeth H Schneider
- Department Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anna N Bukiya
- Department Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Alex M Dopico
- Department Pharmacology, Addiction Science, and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
12
|
Slayden AV, Dyer CL, Ma D, Li W, Bukiya AN, Parrill AL, Dopico AM. Discovery of agonist-antagonist pairs for the modulation of Ca [2]+ and voltage-gated K + channels of large conductance that contain beta1 subunits. Bioorg Med Chem 2022; 68:116876. [PMID: 35716586 PMCID: PMC10464842 DOI: 10.1016/j.bmc.2022.116876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 11/02/2022]
Abstract
Large conductance, calcium/voltage-gated potassium channels (BK) regulate critical body processes, including neuronal, secretory and smooth muscle (SM) function. While BK-forming alpha subunits are ubiquitous, accessory beta1 subunits are highly expressed in SM. This makes beta1 an attractive target for pharmaceutical development to treat SM disorders, such as hypertension or cerebrovascular spasm. Compounds activating BK via beta1 have been identified, yet they exhibit low potency and off-target effects while antagonists that limit agonist activity via beta 1 remain unexplored. Beta1-dependent BK ligand-based pharmacophore modeling and ZINC database searches identified 15 commercially available hits. Concentration-response curves on BK alpha + beta1 subunit-mediated currents were obtained in CHO cells. One potent (EC50 = 20 nM) and highly efficacious activator (maximal activation = ×10.3 of control) was identified along with a potent antagonist (KB = 3.02 nM), both of which were dependent on beta1. Our study provides the first proof-of-principle that an agonist/antagonist pair can be used to control beta1-containing BK activity.
Collapse
Affiliation(s)
- Alexandria V Slayden
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis TN, 38103, USA
| | - Christy L Dyer
- Department of Chemistry, The University of Memphis, Memphis TN, 38152, USA
| | - Dejian Ma
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis TN, 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis TN, 38163, USA
| | - Anna N Bukiya
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis TN, 38103, USA
| | - Abby L Parrill
- Department of Chemistry, The University of Memphis, Memphis TN, 38152, USA
| | - Alex M Dopico
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis TN, 38103, USA.
| |
Collapse
|
13
|
Imaizumi Y. Reciprocal Relationship between Ca 2+ Signaling and Ca 2+-Gated Ion Channels as a Potential Target for Drug Discovery. Biol Pharm Bull 2022; 45:1-18. [PMID: 34980771 DOI: 10.1248/bpb.b21-00896] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cellular Ca2+ signaling functions as one of the most common second messengers of various signal transduction pathways in cells and mediates a number of physiological roles in a cell-type dependent manner. Ca2+ signaling also regulates more general and fundamental cellular activities, including cell proliferation and apoptosis. Among ion channels, Ca2+-permeable channels in the plasma membrane as well as endo- and sarcoplasmic reticulum membranes play important roles in Ca2+ signaling by directly contributing to the influx of Ca2+ from extracellular spaces or its release from storage sites, respectively. Furthermore, Ca2+-gated ion channels in the plasma membrane often crosstalk reciprocally with Ca2+ signals and are central to the regulation of cellular functions. This review focuses on the physiological and pharmacological impact of i) Ca2+-gated ion channels as an apparatus for the conversion of cellular Ca2+ signals to intercellularly propagative electrical signals and ii) the opposite feedback regulation of Ca2+ signaling by Ca2+-gated ion channel activities in excitable and non-excitable cells.
Collapse
Affiliation(s)
- Yuji Imaizumi
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| |
Collapse
|
14
|
Sancho M, Kyle BD. The Large-Conductance, Calcium-Activated Potassium Channel: A Big Key Regulator of Cell Physiology. Front Physiol 2021; 12:750615. [PMID: 34744788 PMCID: PMC8567177 DOI: 10.3389/fphys.2021.750615] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/29/2021] [Indexed: 12/01/2022] Open
Abstract
Large-conductance Ca2+-activated K+ channels facilitate the efflux of K+ ions from a variety of cells and tissues following channel activation. It is now recognized that BK channels undergo a wide range of pre- and post-translational modifications that can dramatically alter their properties and function. This has downstream consequences in affecting cell and tissue excitability, and therefore, function. While finding the “silver bullet” in terms of clinical therapy has remained elusive, ongoing research is providing an impressive range of viable candidate proteins and mechanisms that associate with and modulate BK channel activity, respectively. Here, we provide the hallmarks of BK channel structure and function generally, and discuss important milestones in the efforts to further elucidate the diverse properties of BK channels in its many forms.
Collapse
Affiliation(s)
- Maria Sancho
- Department of Pharmacology, University of Vermont, Burlington, VT, United States
| | - Barry D Kyle
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
15
|
North KC, Bukiya AN, Dopico AM. BK channel-forming slo1 proteins mediate the brain artery constriction evoked by the neurosteroid pregnenolone. Neuropharmacology 2021; 192:108603. [PMID: 34023335 PMCID: PMC8274572 DOI: 10.1016/j.neuropharm.2021.108603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/15/2021] [Accepted: 04/29/2021] [Indexed: 01/24/2023]
Abstract
Pregnenolone is a neurosteroid that modulates glial growth and differentiation, neuronal firing, and several brain functions, these effects being attributed to pregnenolone actions on the neurons and glial cells themselves. Despite the vital role of the cerebral circulation for brain function and the fact that pregnenolone is a vasoactive agent, pregnenolone action on brain arteries remain unknown. Here, we obtained in vivo concentration response curves to pregnenolone on middle cerebral artery (MCA) diameter in anesthetized male and female C57BL/6J mice. In both male and female animals, pregnenolone (1 nM-100 μM) constricted MCA in a concentration-dependent manner, its maximal effect reaching ~22-35% decrease in diameter. Pregnenolone action was replicated in intact and de-endothelialized, in vitro pressurized MCA segments with pregnenolone evoking similar constriction in intact and de-endothelialized MCA. Neurosteroid action was abolished by 1 μM paxilline, a selective blocker of Ca2+ - and voltage-gated K+ channels of large conductance (BK). Cell-attached, patch-clamp recordings on freshly isolated smooth muscle cells from mouse MCAs demonstrated that pregnenolone at concentrations that constricted MCAs in vitro and in vivo (10 μM), reduced BK activity (NPo), with an average decrease in NPo reaching 24.2%. The concentration-dependence of pregnenolone constriction of brain arteries and inhibition of BK activity in intact cells were paralleled by data obtained in cell-free, inside-out patches, with maximal inhibition reached at 10 μM pregnenolone. MCA smooth muscle BKs include channel-forming α (slo1 proteins) and regulatory β1 subunits, encoded by KCNMA1 and KCNMB1, respectively. However, pregnenolone-driven decrease in NPo was still evident in MCA myocytes from KCNMB1-/- mice. Following reconstitution of slo1 channels into artificial, binary phospholipid bilayers, 10 μM pregnenolone evoked slo1 NPo inhibition which was similar to that seen in native membranes. Lastly, pregnenolone failed to constrict MCA from KCNMA1-/- mice. In conclusion, pregnenolone constricts MCA independently of neuronal, glial, endothelial and circulating factors, as well as of cell integrity, organelles, complex membrane cytoarchitecture, and the continuous presence of cytosolic signals. Rather, this action involves direct inhibition of SM BK channels, which does not require β1 subunits but is mediated through direct sensing of the neurosteroid by the channel-forming α subunit.
Collapse
Affiliation(s)
- Kelsey C North
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Anna N Bukiya
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA
| | - Alex M Dopico
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38103, USA.
| |
Collapse
|
16
|
Granados ST, Latorre R, Torres YP. The Membrane Cholesterol Modulates the Interaction Between 17-βEstradiol and the BK Channel. Front Pharmacol 2021; 12:687360. [PMID: 34177597 PMCID: PMC8226216 DOI: 10.3389/fphar.2021.687360] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/24/2021] [Indexed: 11/13/2022] Open
Abstract
BK channels are composed by the pore forming α subunit and, in some tissues, is associated with different accessory β subunits. These proteins modify the biophysical properties of the channel, amplifying the range of BK channel activation according to the physiological context. In the vascular cells, the pore forming BKα subunit is expressed with the β1 subunit, where they play an essential role in the modulation of arterial tone and blood pressure. In eukaryotes, cholesterol is a structural lipid of the cellular membrane. Changes in the ratio of cholesterol content in the plasma membrane (PM) regulates the BK channel activation altering its open probability, and hence, vascular contraction. It has been shown that the estrogen 17β-Estradiol (E2) causes a vasodilator effect in vascular cells, inducing a leftward shift in the V0.5 of the GV curve. Here, we evaluate whether changes in the membrane cholesterol concentration modify the effect that E2 induces on the BKα/β1 channel activity. Using binding and electrophysiology assays after cholesterol depletion or enrichment, we show that the cholesterol enrichment significantly decreases the expression of the α subunit, while cholesterol depletion increased the expression of that α subunit. Additionally, we demonstrated that changes in the membrane cholesterol cause the loss of the modulatory effect of E2 on the BKα/β1 channel activity, without affecting the E2 binding to the complex. Our data suggest that changes in membrane cholesterol content could affect channel properties related to the E2 effect on BKα/β1 channel activity. Finally, the results suggest that an optimal membrane cholesterol content is essential for the activation of BK channels through the β1 subunit.
Collapse
Affiliation(s)
- Sara T Granados
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia.,Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Ramon Latorre
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Yolima P Torres
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
17
|
Martín P, Moncada M, Castillo K, Orsi F, Ducca G, Fernández-Fernández JM, González C, Milesi V. Arachidonic acid effect on the allosteric gating mechanism of BK (Slo1) channels associated with the β1 subunit. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2021; 1863:183550. [PMID: 33417967 DOI: 10.1016/j.bbamem.2021.183550] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/04/2020] [Accepted: 12/30/2020] [Indexed: 11/30/2022]
Abstract
Arachidonic acid (AA) is a fatty acid involved in the modulation of several ion channels. Previously, we reported that AA activates the high conductance Ca2+- and voltage-dependent K+ channel (BK) in vascular smooth muscle depending on the expression of the auxiliary β1 subunit. Here, using the patch-clamp technique on BK channel co-expressed with β1 subunit in a heterologous cell expression system, we analyzed whether AA modifies the three functional modules involved in the channel gating: the voltage sensor domain (VSD), the pore domain (PD), and the intracellular calcium sensor domain (CSD). We present evidence that AA activates BK channel in a direct way, inducing VSD stabilization on its active configuration observed as a significant left shift in the Q-V curve obtained from gating currents recordings. Moreover, AA facilitates the channel opening transitions when VSD are at rest, and the CSD are unoccupied. Furthermore, the activation was independent of the intracellular Ca2+ concentration and reduced when the BK channel was co-expressed with the Y74A mutant of the β1 subunit. These results allow us to present new insigths in the mechanism by which AA modulates BK channels co-expressed with its auxiliary β1 subunit.
Collapse
Affiliation(s)
- Pedro Martín
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, La Plata, Argentina.
| | - Melisa Moncada
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, La Plata, Argentina.
| | - Karen Castillo
- CINV: Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.
| | - Federico Orsi
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, La Plata, Argentina.
| | - Gerónimo Ducca
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, La Plata, Argentina.
| | - José Manuel Fernández-Fernández
- Laboratory of Molecular Physiology, Department of Experimental and Health Sciences, University Pompeu Fabra, 08003 Barcelona, Spain.
| | - Carlos González
- CINV: Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.
| | - Verónica Milesi
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, asociado CIC PBA, Facultad de Ciencias Exactas, La Plata, Argentina.
| |
Collapse
|
18
|
Liu X, Tajima N, Taniguchi M, Kato N. The enantiomer pair of 24S- and 24R-hydroxycholesterol differentially alter activity of large-conductance Ca 2+ -dependent K + (slo1 BK) channel. Chirality 2019; 32:223-230. [PMID: 31756018 DOI: 10.1002/chir.23157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/19/2019] [Accepted: 11/14/2019] [Indexed: 12/20/2022]
Abstract
24S-hydroxycholesterol (HC) is most abundant oxysterols in the brain, passes through blood brain barrier, and is therefore regarded as an intermediary for brain cholesterol elimination. We reported that large-conductance Ca2+ - and voltage-activated K+ (slo1 BK) channels are suppressed by this oxysterol, which is presumably intercalated into cell membrane to access the outer surface of the channel. Such an outer approach would make it difficult to interact with the inner, ion-conducting part of the channel. The present findings showed that 24R-HC, the racemic counterpart of 24S-HC, also suppressed slo1 BK channel but in a different voltage-dependent manner. There was a difference between the effects of the two enantiomers on activation kinetics but not on deactivation kinetics. It is suggested that the chirality contributes to the efficacy of channel blockers that act from outer lipophilic parts of channels, as with those which act on the inner, ion-permeable surface.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Department of Physiology, Kanazawa Medical University, Uchinada, Japan.,Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nobuyoshi Tajima
- Department of Physiology, Kanazawa Medical University, Uchinada, Japan
| | - Makoto Taniguchi
- Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| | - Nobuo Kato
- Department of Physiology, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
19
|
24S-hydroxycholesterol alters activity of large-conductance Ca 2+-dependent K + (slo1 BK) channel through intercalation into plasma membrane. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1525-1535. [PMID: 31136842 DOI: 10.1016/j.bbalip.2019.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 05/11/2019] [Accepted: 05/20/2019] [Indexed: 11/24/2022]
Abstract
Oxysterols, oxidization products of cholesterol, are regarded as bioactive lipids affecting various physiological functions. However, little is known of their effects on ion channels. Using inside-out patch clamp recording, we found that naturally occurring side-chain oxidized oxysterols, 20S‑hydroxycholesterol, 22R‑hydroxycholesterol, 24S‑hydroxycholestero, 25‑hydroxycholesterol, and 27‑hydroxycholesterol, induced current reduction of large-conductance Ca2+- and voltage-activated K+ (slo1 BK) channels heterologously expressed in HEK293T cells. In contrast with side-chain oxidized oxysterols, naturally occurring ring oxidized ones, 7α‑hydroxycholesterol and 7‑ketocholesterol were without effect. By using 24S‑hydroxycholesterol (24S‑HC), the major brain oxysterol, we explored the inhibition mechanism. 24S‑HC inhibited Slo1 BK channels with an IC50 of ~2 μM, and decreased macroscopic current by ~60%. This marked current decrease was accompanied by a rightward shift in the conductance-voltage relationship and a slowed activation kinetics, with the deactivation kinetics unaltered. Furthermore, the membrane sterol scavenger γ‑cyclodextrin was found to rescue slo1 BK channels from the inhibition, implicating that 24S-HC may be intercalated into the plasma membrane to affect the channel. These findings unveil a novel physiological importance of oxysterols from a new angle that involves ion channel regulation.
Collapse
|
20
|
Phospholipid effects on SGLT1-mediated glucose transport in rabbit ileum brush border membrane vesicles. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:182985. [PMID: 31082355 DOI: 10.1016/j.bbamem.2019.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 04/23/2019] [Accepted: 05/08/2019] [Indexed: 11/20/2022]
Abstract
In small intestine, sodium-glucose cotransporter SGLT1 provides the main mechanism for sugar uptake. We investigated the effect of membrane phospholipids (PL) on this transport in rabbit ileal brush border membrane vesicles (BBMV). For this, PL of different charge, length, and saturation were incorporated into BBMV. Transport was measured related to (i) membrane surface charge (membrane-bound MC540 fluorescence), (ii) membrane thickness (PL incorporation of different acyl chain length), and (iii) membrane fluidity (r12AS, fluorescence anisotropy of 12-AS). Compared to phosphatidylcholine (PC) carrying a neutral head group, inhibition of SGLT1 increased considerably with the acidic phosphatidic acid (PA) and phosphatidylinositol (PI) that increase membrane negative surface charge. The order of PL potency was PI>PA > PE = PS > PC. Inhibition by acidic PA-oleate was 5-times more effective than with neutral PE (phosphatidylethanolamine)-oleate. Lineweaver-Burk plot indicated uncompetitive inhibition of SGLT1 by PA. When membrane thickness was increased by neutral PC of varying acyl chain length, transport was increasingly inhibited by 16:1 PC to 22:1 PC. Even more pronounced inhibition was observed with mono-unsaturated instead of saturated acyl chains which increased membrane fluidity (indicated by decreased r12AS). In conclusion, sodium-dependent glucose transport of rabbit ileal BBMV is modulated by (i) altered membrane surface charge, (ii) length of acyl chains via membrane thickness, and (iii) saturation of PL acyl chains altering membrane fluidity. Transport was attenuated by charged PL with longer and unsaturated acyl residues. Alterations of PL may provide a principle for attenuating dietary glucose uptake.
Collapse
|
21
|
Dopico AM, Bukiya AN, Jaggar JH. Calcium- and voltage-gated BK channels in vascular smooth muscle. Pflugers Arch 2018; 470:1271-1289. [PMID: 29748711 DOI: 10.1007/s00424-018-2151-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 04/27/2018] [Indexed: 02/04/2023]
Abstract
Ion channels in vascular smooth muscle regulate myogenic tone and vessel contractility. In particular, activation of calcium- and voltage-gated potassium channels of large conductance (BK channels) results in outward current that shifts the membrane potential toward more negative values, triggering a negative feed-back loop on depolarization-induced calcium influx and SM contraction. In this short review, we first present the molecular basis of vascular smooth muscle BK channels and the role of subunit composition and trafficking in the regulation of myogenic tone and vascular contractility. BK channel modulation by endogenous signaling molecules, and paracrine and endocrine mediators follows. Lastly, we describe the functional changes in smooth muscle BK channels that contribute to, or are triggered by, common physiological conditions and pathologies, including obesity, diabetes, and systemic hypertension.
Collapse
Affiliation(s)
- Alex M Dopico
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, 71 South Manassas St., Memphis, TN, 38163, USA.
| | - Anna N Bukiya
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, 71 South Manassas St., Memphis, TN, 38163, USA
| | - Jonathan H Jaggar
- Department of Physiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
22
|
Simakova MN, Bisen S, Dopico AM, Bukiya AN. Statin therapy exacerbates alcohol-induced constriction of cerebral arteries via modulation of ethanol-induced BK channel inhibition in vascular smooth muscle. Biochem Pharmacol 2017; 145:81-93. [PMID: 28865873 DOI: 10.1016/j.bcp.2017.08.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/28/2017] [Indexed: 12/26/2022]
Abstract
Statins constitute the most commonly prescribed drugs to decrease cholesterol (CLR). CLR is an important modulator of alcohol-induced cerebral artery constriction (AICAC). Using rats on a high CLR diet (2% CLR) we set to determine whether atorvastatin administration (10mg/kg daily for 18-23weeks) modified AICAC. Middle cerebral arteries were pressurized in vitro at 60mmHg and AICAC was evoked by 50mM ethanol, that is within the range of blood alcohol detected in humans following moderate-to-heavy drinking. AICAC was evident in high CLR+atorvastatin group but not in high CLR diet+placebo. Statin exacerbation of AICAC persisted in de-endothelialized arteries, and was blunted by CLR enrichment in vitro. Fluorescence imaging of filipin-stained arteries showed that atorvastatin decreased vascular smooth muscle (VSM) CLR when compared to placebo, this difference being reduced by CLR enrichment in vitro. Voltage- and calcium-gated potassium channels of large conductance (BK) are known VSM targets of ethanol, with their beta1 subunit being necessary for ethanol-induced channel inhibition and resulting AICAC. Ethanol-induced BK inhibition in excised membrane patches from freshly isolated myocytes was exacerbated in the high CLR diet+atorvastatin group when compared to high CLR diet+placebo. Unexpectedly, atorvastatin decreased the amount and function of BK beta1 subunit as documented by immunofluorescence imaging and functional patch-clamp studies. Atorvastatin exacerbation of ethanol-induced BK inhibition disappeared upon artery CLR enrichment in vitro. Our study demonstrates for the first time statin's ability to exacerbate the vascular effect of a widely consumed drug of abuse, this exacerbation being driven by statin modulation of ethanol-induced BK channel inhibition in the VSM via CLR-mediated mechanism.
Collapse
Affiliation(s)
- Maria N Simakova
- Dept. Pharmacology, University of Tennessee HSC, Memphis, TN 38103, United States
| | - Shivantika Bisen
- Dept. Pharmacology, University of Tennessee HSC, Memphis, TN 38103, United States
| | - Alex M Dopico
- Dept. Pharmacology, University of Tennessee HSC, Memphis, TN 38103, United States
| | - Anna N Bukiya
- Dept. Pharmacology, University of Tennessee HSC, Memphis, TN 38103, United States.
| |
Collapse
|
23
|
Dopico AM, Bukiya AN. Regulation of Ca 2+-Sensitive K + Channels by Cholesterol and Bile Acids via Distinct Channel Subunits and Sites. CURRENT TOPICS IN MEMBRANES 2017; 80:53-93. [PMID: 28863822 DOI: 10.1016/bs.ctm.2017.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cholesterol (CLR) conversion into bile acids (BAs) in the liver constitutes the major pathway for CLR elimination from the body. Moreover, these steroids regulate each other's metabolism. While the roles of CLR and BAs in regulating metabolism and tissue function are well known, research of the last two decades revealed the existence of specific protein receptors for CLR or BAs in tissues with minor contribution to lipid metabolism, raising the possibility that these lipids serve as signaling molecules throughout the body. Among other lipids, CLR and BAs regulate ionic current mediated by the activity of voltage- and Ca2+-gated, K+ channels of large conductance (BK channels) and, thus, modulate cell physiology and participate in tissue pathophysiology. Initial work attributed modification of BK channel function by CLR or BAs to the capability of these steroids to directly interact with bilayer lipids and thus alter the physicochemical properties of the bilayer with eventual modification of BK channel function. Based on our own work and that of others, we now review evidence that supports direct interactions between CLR or BA and specific BK protein subunits, and the consequence of such interactions on channel activity and organ function, with a particular emphasis on arterial smooth muscle. For each steroid type, we will also briefly discuss several mechanisms that may underlie modification of channel steady-state activity. Finally, we will present novel computational data that provide a chemical basis for differential recognition of CLR vs lithocholic acid by distinct BK channel subunits and recognition sites.
Collapse
Affiliation(s)
- Alex M Dopico
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States.
| | - Anna N Bukiya
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
24
|
Alcohol Regulates BK Surface Expression via Wnt/β-Catenin Signaling. J Neurosci 2017; 36:10625-10639. [PMID: 27733613 DOI: 10.1523/jneurosci.0491-16.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 07/27/2016] [Indexed: 12/26/2022] Open
Abstract
It has been suggested that drug tolerance represents a form of learning and memory, but this has not been experimentally established at the molecular level. We show that a component of alcohol molecular tolerance (channel internalization) from rat hippocampal neurons requires protein synthesis, in common with other forms of learning and memory. We identify β-catenin as a primary necessary protein. Alcohol increases β-catenin, and blocking accumulation of β-catenin blocks alcohol-induced internalization in these neurons. In transfected HEK293 cells, suppression of Wnt/β-catenin signaling blocks ethanol-induced internalization. Conversely, activation of Wnt/β-catenin reduces BK current density. A point mutation in a putative glycogen synthase kinase phosophorylation site within the S10 region of BK blocks internalization, suggesting that Wnt/β-catenin directly regulates alcohol-induced BK internalization via glycogen synthase kinase phosphorylation. These findings establish de novo protein synthesis and Wnt/β-catenin signaling as critical in mediating a persistent form of BK molecular alcohol tolerance establishing a commonality with other forms of long-term plasticity. SIGNIFICANCE STATEMENT Alcohol tolerance is a key step toward escalating alcohol consumption and subsequent dependence. Our research aims to make significant contributions toward novel, therapeutic approaches to prevent and treat alcohol misuse by understanding the molecular mechanisms of alcohol tolerance. In our current study, we identify the role of a key regulatory pathway in alcohol-induced persistent molecular changes within the hippocampus. The canonical Wnt/β-catenin pathway regulates BK channel surface expression in a protein synthesis-dependent manner reminiscent of other forms of long-term hippocampal neuronal adaptations. This unique insight opens the possibility of using clinically tested drugs, targeting the Wnt/β-catenin pathway, for the novel use of preventing and treating alcohol dependency.
Collapse
|
25
|
Effect of Statins on the Nanomechanical Properties of Supported Lipid Bilayers. Biophys J 2017; 111:363-372. [PMID: 27463138 DOI: 10.1016/j.bpj.2016.06.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 05/25/2016] [Accepted: 06/15/2016] [Indexed: 11/23/2022] Open
Abstract
Many drugs and other xenobiotics may reach systemic concentrations where they interact not only with the proteins that are their therapeutic targets but also modify the physicochemical properties of the cell membrane, which may lead to altered function of many transmembrane proteins beyond the intended targets. These changes in bilayer properties may contribute to nonspecific, promiscuous changes in membrane protein and cell function because membrane proteins are energetically coupled to their host lipid bilayer. It is thus important, for both pharmaceutical and biophysical reasons, to understand the bilayer-modifying effect of amphiphiles (including therapeutic agents). Here we use atomic force microscopy topography imaging and nanomechanical mapping to monitor the effect of statins, a family of hypolipidemic drugs, on synthetic lipid membranes. Our results reveal that statins alter the nanomechanical stability of the bilayers and increase their elastic moduli depending on the lipid bilayer order. Our results also suggest that statins increase bilayer heterogeneity, which may indicate that statins form nanometer-sized aggregates in the membrane. This is further evidence that changes in bilayer nanoscale mechanical properties may be a signature of lipid bilayer-mediated effects of amphiphilic drugs.
Collapse
|
26
|
Di Scala C, Baier CJ, Evans LS, Williamson PT, Fantini J, Barrantes FJ. Relevance of CARC and CRAC Cholesterol-Recognition Motifs in the Nicotinic Acetylcholine Receptor and Other Membrane-Bound Receptors. CURRENT TOPICS IN MEMBRANES 2017; 80:3-23. [DOI: 10.1016/bs.ctm.2017.05.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
27
|
Kuntamallappanavar G, Bisen S, Bukiya AN, Dopico AM. Differential distribution and functional impact of BK channel beta1 subunits across mesenteric, coronary, and different cerebral arteries of the rat. Pflugers Arch 2016; 469:263-277. [PMID: 28012000 DOI: 10.1007/s00424-016-1929-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/06/2016] [Accepted: 12/12/2016] [Indexed: 12/12/2022]
Abstract
Large conductance, Ca2+i- and voltage-gated K+ (BK) channels regulate myogenic tone and, thus, arterial diameter. In smooth muscle (SM), BK channels include channel-forming α and auxiliary β1 subunits. BK β1 increases the channel's Ca2+ sensitivity, allowing BK channels to negatively feedback on depolarization-induced Ca2+ entry, oppose SM contraction and favor vasodilation. Thus, endothelial-independent vasodilation can be evoked though targeting of SM BK β1 by endogenous ligands, including lithocholate (LCA). Here, we investigated the expression of BK β1 across arteries of the cerebral and peripheral circulations, and the contribution of such expression to channel function and BK β1-mediated vasodilation. Data demonstrate that endothelium-independent, BK β1-mediated vasodilation by LCA is larger in coronary (CA) and basilar (BA) arteries than in anterior cerebral (ACA), middle cerebral (MCA), posterior cerebral (PCA), and mesenteric (MA) arteries, all arterial segments having a similar diameter. Thus, differential dilation occurs in extracranial arteries which are subjected to similar vascular pressure (CA vs. MA) and in arteries that irrigate different brain regions (BA vs. ACA, MCA, and PCA). SM BK channels from BA and CA displayed increased basal activity and LCA responses, indicating increased BK β1 functional presence. Indeed, in the absence of detectable changes in BK α, BA and CA myocytes showed an increased location of BK β1 in the plasmalemma/subplasmalemma. Moreover, these myocytes distinctly showed increased BK β1 messenger RNA (mRNA) levels. Supporting a major role of enhanced BK β1 transcripts in artery dilation, LCA-induced dilation of MCA transfected with BK β1 complementary DNA (cDNA) was as high as LCA-induced dilation of untransfected BA or CA.
Collapse
Affiliation(s)
- Guruprasad Kuntamallappanavar
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, 71 South Manassas St, Memphis, TN, 38103, USA
| | - Shivantika Bisen
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, 71 South Manassas St, Memphis, TN, 38103, USA
| | - Anna N Bukiya
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, 71 South Manassas St, Memphis, TN, 38103, USA
| | - Alex M Dopico
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, 71 South Manassas St, Memphis, TN, 38103, USA.
| |
Collapse
|
28
|
Cheng X, Tan X, Liu W, Li H, Yan L, Yang Y, Zeng X, Cao J. PI4Kβ, PIPs and BK Ca channel function. Sci Bull (Beijing) 2016. [DOI: 10.1007/s11434-016-1186-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
29
|
Illison J, Tian L, McClafferty H, Werno M, Chamberlain LH, Leiss V, Sassmann A, Offermanns S, Ruth P, Shipston MJ, Lukowski R. Obesogenic and Diabetogenic Effects of High-Calorie Nutrition Require Adipocyte BK Channels. Diabetes 2016; 65:3621-3635. [PMID: 27605626 DOI: 10.2337/db16-0245] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 08/16/2016] [Indexed: 11/13/2022]
Abstract
Elevated adipose tissue expression of the Ca2+- and voltage-activated K+ (BK) channel was identified in morbidly obese men carrying a BK gene variant, supporting the hypothesis that K+ channels affect the metabolic responses of fat cells to nutrients. To establish the role of endogenous BKs in fat cell maturation, storage of excess dietary fat, and body weight (BW) gain, we studied a gene-targeted mouse model with global ablation of the BK channel (BKL1/L1) and adipocyte-specific BK-deficient (adipoqBKL1/L2) mice. Global BK deficiency afforded protection from BW gain and excessive fat accumulation induced by a high-fat diet (HFD). Expansion of white adipose tissue-derived epididymal BKL1/L1 preadipocytes and their differentiation to lipid-filled mature adipocytes in vitro, however, were improved. Moreover, BW gain and total fat masses of usually superobese ob/ob mice were significantly attenuated in the absence of BK, together supporting a central or peripheral role for BKs in the regulatory system that controls adipose tissue and weight. Accordingly, HFD-fed adipoqBKL1/L2 mutant mice presented with a reduced total BW and overall body fat mass, smaller adipocytes, and reduced leptin levels. Protection from pathological weight gain in the absence of adipocyte BKs was beneficial for glucose handling and related to an increase in body core temperature as a result of higher levels of uncoupling protein 1 and a low abundance of the proinflammatory interleukin-6, a common risk factor for diabetes and metabolic abnormalities. This suggests that adipocyte BK activity is at least partially responsible for excessive BW gain under high-calorie conditions, suggesting that BK channels are promising drug targets for pharmacotherapy of metabolic disorders and obesity.
Collapse
Affiliation(s)
- Julia Illison
- Pharmakologie, Toxikologie und Klinische Pharmazie, Institut für Pharmazie, Tübingen, Germany
| | - Lijun Tian
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, U.K
| | - Heather McClafferty
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, U.K
| | - Martin Werno
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Strathclyde University, Glasgow, U.K
| | - Luke H Chamberlain
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Strathclyde University, Glasgow, U.K
| | - Veronika Leiss
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, University Hospital Tübingen, Tübingen, Germany
| | - Antonia Sassmann
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Peter Ruth
- Pharmakologie, Toxikologie und Klinische Pharmazie, Institut für Pharmazie, Tübingen, Germany
| | - Michael J Shipston
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, U.K
| | - Robert Lukowski
- Pharmakologie, Toxikologie und Klinische Pharmazie, Institut für Pharmazie, Tübingen, Germany
| |
Collapse
|
30
|
Stearoyl-CoA desaturase-1 and adaptive stress signaling. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1719-1726. [DOI: 10.1016/j.bbalip.2016.08.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/09/2016] [Accepted: 08/17/2016] [Indexed: 12/31/2022]
|
31
|
Hoshi T, Heinemann SH. Modulation of BK Channels by Small Endogenous Molecules and Pharmaceutical Channel Openers. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 128:193-237. [PMID: 27238265 DOI: 10.1016/bs.irn.2016.03.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Voltage- and Ca(2+)-activated K(+) channels of big conductance (BK channels) are abundantly found in various organs and their relevance for smooth muscle tone and neuronal signaling is well documented. Dysfunction of BK channels is implicated in an array of human diseases involving many organs including the nervous, pulmonary, cardiovascular, renal, and urinary systems. In humans a single gene (KCNMA1) encodes the pore-forming α subunit (Slo1) of BK channels, but the channel properties are variable because of alternative splicing, tissue- and subcellular-specific auxiliary subunits (β, γ), posttranslational modifications, and a multitude of endogenous signaling molecules directly affecting the channel function. Initiatives to develop drugs capable of activating BK channels (channel openers) therefore need to consider the tissue-specific variability of BK channel structure and the potential interference with endogenously produced regulatory factors. The atomic structural basis of BK channel function is only beginning to be revealed. However, building on detailed knowledge of BK channel function, including its single-channel characteristics, voltage- and Ca(2+) dependence of channel gating, and modulation by diffusible messengers, a multi-tier allosteric model of BK channel gating (Horrigan and Aldrich (HA) model) has become a valuable tool in studying modulation of the channel. Using the conceptual framework of the HA model, we here review the functional impact of endogenous modulatory factors and select small synthetic compounds that regulate BK channel activity. Furthermore, we devise experimental approaches for studying BK channel-drug interactions with the aim to classify BK-modulating substances according to their molecular mode of action.
Collapse
Affiliation(s)
- T Hoshi
- University of Pennsylvania, Philadelphia, PA, United States.
| | - S H Heinemann
- Friedrich Schiller University Jena & Jena University Hospital, Jena, Germany
| |
Collapse
|
32
|
Krishnamoorthy-Natarajan G, Koide M. BK Channels in the Vascular System. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 128:401-38. [PMID: 27238270 DOI: 10.1016/bs.irn.2016.03.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autoregulation of blood flow is essential for the preservation of organ function to ensure continuous supply of oxygen and essential nutrients and removal of metabolic waste. This is achieved by controlling the diameter of muscular arteries and arterioles that exhibit a myogenic response to changes in arterial blood pressure, nerve activity and tissue metabolism. Large-conductance voltage and Ca(2+)-dependent K(+) channels (BK channels), expressed exclusively in smooth muscle cells (SMCs) in the vascular wall of healthy arteries, play a critical role in regulating the myogenic response. Activation of BK channels by intracellular, local, and transient ryanodine receptor-mediated "Ca(2+) sparks," provides a hyperpolarizing influence on the SMC membrane potential thereby decreasing the activity of voltage-dependent Ca(2+) channels and limiting Ca(2+) influx to promote SMC relaxation and vasodilation. The BK channel α subunit, a large tetrameric protein with each monomer consisting of seven-transmembrane domains, a long intracellular C-terminal tail and an extracellular N-terminus, associates with the β1 and γ subunits in vascular SMCs. The BK channel is regulated by factors originating within the SMC or from the endothelium, perivascular nerves and circulating blood, that significantly alter channel gating properties, Ca(2+) sensitivity and expression of the α and/or β1 subunit. The BK channel thus serves as a central receiving dock that relays the effects of the changes in several such concomitant autocrine and paracrine factors and influences cardiovascular health. This chapter describes the primary mechanism of regulation of myogenic response by BK channels and the alterations to this mechanism wrought by different vasoactive mediators.
Collapse
Affiliation(s)
| | - M Koide
- University of Vermont, Burlington, VT, United States
| |
Collapse
|
33
|
Abstract
BK channels are universal regulators of cell excitability, given their exceptional unitary conductance selective for K(+), joint activation mechanism by membrane depolarization and intracellular [Ca(2+)] elevation, and broad expression pattern. In this chapter, we discuss the structural basis and operational principles of their activation, or gating, by membrane potential and calcium. We also discuss how the two activation mechanisms interact to culminate in channel opening. As members of the voltage-gated potassium channel superfamily, BK channels are discussed in the context of archetypal family members, in terms of similarities that help us understand their function, but also seminal structural and biophysical differences that confer unique functional properties.
Collapse
Affiliation(s)
- A Pantazis
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States
| | - R Olcese
- David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, United States.
| |
Collapse
|
34
|
Yu M, Liu SL, Sun PB, Pan H, Tian CL, Zhang LH. Peptide toxins and small-molecule blockers of BK channels. Acta Pharmacol Sin 2016; 37:56-66. [PMID: 26725735 DOI: 10.1038/aps.2015.139] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/13/2015] [Indexed: 12/21/2022]
Abstract
Large conductance, Ca(2+)-activated potassium (BK) channels play important roles in the regulation of neuronal excitability and the control of smooth muscle contractions. BK channels can be activated by changes in both the membrane potential and intracellular Ca(2+) concentrations. Here, we provide an overview of the structural and pharmacological properties of BK channel blockers. First, the properties of different venom peptide toxins from scorpions and snakes are described, with a focus on their characteristic structural motifs, including their disulfide bond formation pattern, the binding interface between the toxin and BK channel, and the functional consequence of the blockage of BK channels by these toxins. Then, some representative non-peptide blockers of BK channels are also described, including their molecular formula and pharmacological effects on BK channels. The detailed categorization and descriptions of these BK channel blockers will provide mechanistic insights into the blockade of BK channels. The structures of peptide toxins and non-peptide compounds could provide templates for the design of new channel blockers, and facilitate the optimization of lead compounds for further therapeutic applications in neurological disorders or cardiovascular diseases.
Collapse
|
35
|
Tian Y, Ullrich F, Xu R, Heinemann SH, Hou S, Hoshi T. Two distinct effects of PIP2 underlie auxiliary subunit-dependent modulation of Slo1 BK channels. ACTA ACUST UNITED AC 2015; 145:331-43. [PMID: 25825171 PMCID: PMC4380209 DOI: 10.1085/jgp.201511363] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Phosphatidylinositol 4,5-bisphosphate (PIP2) plays a critical role in modulating the function of numerous ion channels, including large-conductance Ca(2+)- and voltage-dependent K(+) (BK, Slo1) channels. Slo1 BK channel complexes include four pore-forming Slo1 (α) subunits as well as various regulatory auxiliary subunits (β and γ) that are expressed in different tissues. We examined the molecular and biophysical mechanisms underlying the effects of brain-derived PIP2 on human Slo1 BK channel complexes with different subunit compositions that were heterologously expressed in human embryonic kidney cells. PIP2 inhibited macroscopic currents through Slo1 channels without auxiliary subunits and through Slo1 + γ1 complexes. In contrast, PIP2 markedly increased macroscopic currents through Slo1 + β1 and Slo1 + β4 channel complexes and failed to alter macroscopic currents through Slo1 + β2 and Slo1 + β2 Δ2-19 channel complexes. Results obtained at various membrane potentials and divalent cation concentrations suggest that PIP2 promotes opening of the ion conduction gate in all channel types, regardless of the specific subunit composition. However, in the absence of β subunits positioned near the voltage-sensor domains (VSDs), as in Slo1 and probably Slo1 + γ1, PIP2 augments the negative surface charge on the cytoplasmic side of the membrane, thereby shifting the voltage dependence of VSD-mediated activation in the positive direction. When β1 or β4 subunits occupy the space surrounding the VSDs, only the stimulatory effect of PIP2 is evident. The subunit compositions of native Slo1 BK channels differ in various cell types; thus, PIP2 may exert distinct tissue- and divalent cation-dependent modulatory influences.
Collapse
Affiliation(s)
- Yutao Tian
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104
| | - Florian Ullrich
- Department of Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena and Jena University Hospital, D-07745 Jena, Germany
| | - Rong Xu
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104
| | - Stefan H Heinemann
- Department of Biophysics, Center for Molecular Biomedicine, Friedrich Schiller University Jena and Jena University Hospital, D-07745 Jena, Germany
| | - Shangwei Hou
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Toshinori Hoshi
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
36
|
Spector DA, Deng J, Coleman R, Wade JB. The urothelium of a hibernator: the American black bear. Physiol Rep 2015; 3:e12429. [PMID: 26109187 PMCID: PMC4510630 DOI: 10.14814/phy2.12429] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 05/13/2015] [Accepted: 05/18/2015] [Indexed: 12/27/2022] Open
Abstract
The American black bear undergoes a 3-5 month winter hibernation during which time bears do not eat, drink, defecate, or urinate. During hibernation renal function (GFR) is 16-50% of normal but urine is reabsorbed across the urinary bladder (UB) urothelium thus enabling metabolic recycling of all urinary constituents. To elucidate the mechanism(s) whereby urine is reabsorbed, we examined the UBs of five nonhibernating wild bears using light, electron (EM), and confocal immunofluorescent (IF) microscopy-concentrating on two components of the urothelial permeability barrier - the umbrella cell apical membranes and tight junctions (TJ). Bear UB has the same tissue layers (serosa, muscularis, lamina propria, urothelia) and its urothelia has the same cell layers (basal, intermediate, umbrella cells) as other mammalians. By EM, the bear apical membrane demonstrated a typical mammalian scalloped appearance with hinge and plaque regions - the latter containing an asymmetric trilaminar membrane and, on IF, uroplakins Ia, IIIa, and IIIb. The umbrella cell TJs appeared similar to those in other mammals and also contained TJ proteins occludin and claudin - 4, and not claudin -2. Thus, we were unable to demonstrate urothelial apical membrane or TJ differences between active black bears and other mammals. Expression and localization of UT-B, AQP-1 and -3, and Na(+), K(+)-ATPase on bear urothelial membranes was similar to that of other mammals. Similar studies of urothelia of hibernating bears, including evaluation of the apical membrane lipid bilayer and GAGs layer are warranted to elucidate the mechanism(s) whereby hibernating bears reabsorb their daily urine output and thus ensure successful hibernation.
Collapse
Affiliation(s)
- David A Spector
- Division of Renal Medicine, Johns Hopkins Bayview Medical Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jie Deng
- Division of Renal Medicine, Johns Hopkins Bayview Medical Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Richard Coleman
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - James B Wade
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
37
|
Dopico AM, Bukiya AN, Martin GE. Ethanol modulation of mammalian BK channels in excitable tissues: molecular targets and their possible contribution to alcohol-induced altered behavior. Front Physiol 2014; 5:466. [PMID: 25538625 PMCID: PMC4256990 DOI: 10.3389/fphys.2014.00466] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 11/13/2014] [Indexed: 11/30/2022] Open
Abstract
In most tissues, the function of Ca2+- and voltage-gated K+ (BK) channels is modified in response to ethanol concentrations reached in human blood during alcohol intoxication. In general, modification of BK current from ethanol-naïve preparations in response to brief ethanol exposure results from changes in channel open probability without modification of unitary conductance or change in BK protein levels in the membrane. Protracted and/or repeated ethanol exposure, however, may evoke changes in BK expression. The final ethanol effect on BK open probability leading to either BK current potentiation or BK current reduction is determined by an orchestration of molecular factors, including levels of activating ligand (Ca2+i), BK subunit composition and post-translational modifications, and the channel's lipid microenvironment. These factors seem to allosterically regulate a direct interaction between ethanol and a recognition pocket of discrete dimensions recently mapped to the channel-forming (slo1) subunit. Type of ethanol exposure also plays a role in the final BK response to the drug: in several central nervous system regions (e.g., striatum, primary sensory neurons, and supraoptic nucleus), acute exposure to ethanol reduces neuronal excitability by enhancing BK activity. In contrast, protracted or repetitive ethanol administration may alter BK subunit composition and membrane expression, rendering the BK complex insensitive to further ethanol exposure. In neurohypophyseal axon terminals, ethanol potentiation of BK channel activity leads to a reduction in neuropeptide release. In vascular smooth muscle, however, ethanol inhibition of BK current leads to cell contraction and vascular constriction.
Collapse
Affiliation(s)
- Alex M Dopico
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center Memphis, TN, USA
| | - Anna N Bukiya
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center Memphis, TN, USA
| | - Gilles E Martin
- Department of Psychiatry, The University of Massachusetts Medical School Worcester, MA, USA
| |
Collapse
|
38
|
Bukiya AN, McMillan J, Liu J, Shivakumar B, Parrill AL, Dopico AM. Activation of calcium- and voltage-gated potassium channels of large conductance by leukotriene B4. J Biol Chem 2014; 289:35314-25. [PMID: 25371198 DOI: 10.1074/jbc.m114.577825] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Calcium/voltage-gated, large conductance potassium (BK) channels control numerous physiological processes, including myogenic tone. BK channel regulation by direct interaction between lipid and channel protein sites has received increasing attention. Leukotrienes (LTA4, LTB4, LTC4, LTD4, and LTE4) are inflammatory lipid mediators. We performed patch clamp studies in Xenopus oocytes that co-expressed BK channel-forming (cbv1) and accessory β1 subunits cloned from rat cerebral artery myocytes. Leukotrienes were applied at 0.1 nm-10 μm to either leaflet of cell-free membranes at a wide range of [Ca(2+)]i and voltages. Only LTB4 reversibly increased BK steady-state activity (EC50 = 1 nm; Emax reached at 10 nm), with physiological [Ca(2+)]i and voltages favoring this activation. Homomeric cbv1 or cbv1-β2 channels were LTB4-resistant. Computational modeling predicted that LTB4 docked onto the cholane steroid-sensing site in the BK β1 transmembrane domain 2 (TM2). Co-application of LTB4 and cholane steroid did not further increase LTB4-induced activation. LTB4 failed to activate β1 subunit-containing channels when β1 carried T169A, A176S, or K179I within the docking site. Co-application of LTB4 with LTA4, LTC4, LTD4, or LTE4 suppressed LTB4-induced activation. Inactive leukotrienes docked onto a portion of the site, probably preventing tight docking of LTB4. In summary, we document the ability of two endogenous lipids from different chemical families to share their site of action on a channel accessory subunit. Thus, cross-talk between leukotrienes and cholane steroids might converge on regulation of smooth muscle contractility via BK β1. Moreover, the identification of LTB4 as a highly potent ligand for BK channels is critical for the future development of β1-specific BK channel activators.
Collapse
Affiliation(s)
- Anna N Bukiya
- From the Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Jacob McMillan
- the Department of Chemistry and Computational Research on Materials Institute (CROMIUM), University of Memphis, Memphis, Tennessee 38152
| | - Jianxi Liu
- From the Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Bangalore Shivakumar
- From the Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| | - Abby L Parrill
- the Department of Chemistry and Computational Research on Materials Institute (CROMIUM), University of Memphis, Memphis, Tennessee 38152
| | - Alex M Dopico
- From the Department of Pharmacology, University of Tennessee Health Science Center, Memphis, Tennessee 38163 and
| |
Collapse
|