1
|
Mahmood NMS, Mahmood AMR, Maulood IM. The roles of melatonin and potassium channels in relaxation response to ang 1-7 in diabetic rat isolated aorta. Cytotechnology 2025; 77:55. [PMID: 39927136 PMCID: PMC11799518 DOI: 10.1007/s10616-025-00720-y] [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: 12/18/2024] [Accepted: 01/27/2025] [Indexed: 02/11/2025] Open
Abstract
In a circadian cycle, the pineal gland produces and releases melatonin (MEL) into the bloodstream. By activating distinct melatonin receptors, MEL has been shown to variably change vascular endothelial dysfunction (VED) to various vascular beds. This study investigates the interaction of melatonin (MEL) and potassium ion (K+) on angiotensin 1-7 (Ang 1-7) vasorelaxant in streptozotocin (STZ)-induced diabetes mellitus (DM) and non-diabetes mellitus (non-DM) male albino rat aortic rings. The isometric tension of isolated aortic rings was assessed by generating a dose-response curve (DRC) for Ang 1-7 using a PowerLab data acquisition system. Accordingly, three experimental sets were carried out. In the first set the aortic rings were exposed MEL and MEL agonist ramelteon (RAM) and MEL antagonist luzindole (LUZ). In the second set, the aortic rings were exposed to various non-selective calcium activated potassium channel (KCa) blockers, including tetraethylammonium (TEA), a small and large-conductance calcium-activated K+ [(SKCa) and (BKCa)] channels blocker charybdotoxin (ChTx) and intermediate calcium-activated K+ channel (IKCa) blocker clotrimazole (CLT). In the third set, the aortic rings were exposed to various selective K+ channels blockers, including the selective blocker of KATP channel, glibenclamide (Glib), 4-aminopyridine (4-AP), a selective blocker of Kv channels and BaCl2, delayed inward rectifier K+ channels (Kir) blocker. The results highlight the significant role of MEL in modulating vascular reactivity, particularly in the DM aorta. By enhancing the vasorelaxant effects of Ang 1-7 through mechanisms involving its receptors and antioxidant activities, MEL demonstrates its potential to counteract oxidative stress and VED associated with diabetes. These findings advance the understanding of vascular reactivity in diabetes and suggest MEL as a promising therapeutic agent for improving vascular health in diabetic conditions.
Collapse
Affiliation(s)
- Nazar M. Shareef Mahmood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region Iraq
| | - Almas M. R. Mahmood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region Iraq
| | - Ismail M. Maulood
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region Iraq
| |
Collapse
|
2
|
Vasileva VY, Sudarikova AV, Chubinskiy-Nadezhdin VI. Functional coupling of Piezo1 channels and Ca 2+-activated ion channels in the plasma membrane: fine-tunable interplay with wide-range signaling effects. Am J Physiol Cell Physiol 2025; 328:C1338-C1345. [PMID: 40099870 DOI: 10.1152/ajpcell.00094.2025] [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: 01/31/2025] [Revised: 02/11/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
Ca2+ is a universal second messenger in living cells, and its concentration should be precisely localized to provide the outstanding specificity of signal transduction. The conception of Ca2+ micro- and nanodomains in which Ca2+ ions could control the activity of various Ca2+-dependent molecules was postulated: the Ca2+-permeable ion channels in the plasma membrane provide a pathway for Ca2+ entry from the extracellular milieu into the cytosol regulating the activity of Ca2+-dependent molecules, that is, functionally colocalized Ca2+-activated ion channels. These channel complexes of different molecular compositions were observed in the cells of different origins; thus, the phenomenon of ion channel coupling is thought to be a universal property of living cells. Piezo1 is a mechanosensitive Ca2+-permeable ion channel that plays a pivotal role in cellular mechanotransduction and is integrated into various signaling cascades regulating the activity of Ca2+-dependent molecules. Here, we summarized recent experimental data on the presence and role of functional complexes of Piezo1 with Ca2+-activated channels of different origins and highlighted the complex molecular mechanisms that could control the channel coupling in the plasma membrane.
Collapse
|
3
|
Thammavongsa DA, Jackvony TN, Bookland MJ, Tang-Schomer MD. Targeting Ion Channels: Blockers Suppress Calcium Signals and Induce Cytotoxicity Across Medulloblastoma Cell Models. Bioengineering (Basel) 2025; 12:268. [PMID: 40150732 PMCID: PMC11939613 DOI: 10.3390/bioengineering12030268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/25/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Medulloblastoma (MB) groups 3 and 4 lack targeted therapies despite their dismal prognoses. Ion channels and pumps have been implicated in promoting MB metastasis and growth; however, their roles remain poorly understood. In this study, we repurposed FDA-approved channel blockers and modulators to investigate their potential anti-tumor effects in MB cell lines (DAOY and D283) and primary cell cultures derived from a patient with MB. For the first time, we report spontaneous calcium signaling in MB cells. Spontaneous calcium signals were significantly reduced by mibefradil (calcium channel blocker), paxilline (calcium-activated potassium channel blocker), and thioridazine (potassium channel blocker). These drugs induced dose-dependent cytotoxicity in both the DAOY and D283 cell lines, as well as in primary cell cultures of a patient with group 3 or 4 MB. In contrast, digoxin and ouabain, inhibitors of the Na/K pump, reduced the calcium signaling by over 90% in DAOY cells and induced approximately 90% cell death in DAOY cells and 80% cell death in D283 cells. However, these effects were significantly diminished in the cells derived from a patient with MB, highlighting the variability in drug sensitivity among MB models. These findings demonstrate that calcium signaling is critical for MB cell survival and that the targeted inhibition of calcium pathways suppresses tumor cell growth across multiple MB models.
Collapse
Affiliation(s)
- Darani Ashley Thammavongsa
- UConn Health, Department of Pediatrics, 263 Farmington Avenue, Farmington, CT 06030, USA; (D.A.T.); (T.N.J.)
| | - Taylor N. Jackvony
- UConn Health, Department of Pediatrics, 263 Farmington Avenue, Farmington, CT 06030, USA; (D.A.T.); (T.N.J.)
| | - Markus J. Bookland
- Connecticut Children’s Medical Center, 282 Washington St, Hartford, CT 06106, USA;
| | - Min D. Tang-Schomer
- UConn Health, Department of Pediatrics, 263 Farmington Avenue, Farmington, CT 06030, USA; (D.A.T.); (T.N.J.)
- Connecticut Children’s Medical Center, 282 Washington St, Hartford, CT 06106, USA;
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06030, USA
| |
Collapse
|
4
|
Tang YB, Tang L, Chen B, Fan MJ, Chen GJ, Ou YN, Yang F, Wu XZ. Intranasal oxytocin alleviates postsurgical pain and comorbid anxiety in mice: Participation of BK(Ca) channels in the hippocampus. Neuropharmacology 2025; 265:110243. [PMID: 39631680 DOI: 10.1016/j.neuropharm.2024.110243] [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: 09/02/2024] [Revised: 11/10/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
The affective dimension in postsurgical pain is still poorly understood. Since neuropeptide oxytocin (OXT) has been implicated in a broad spectrum of pain and negative emotion, we investigated the potential therapeutic effect of intranasal OXT on postsurgical pain and associated anxiety in a mice model of plantar incision. The role of large conductance Ca(2+)-activated K(+) (BK(Ca)) channels was explored by using behavioral pharmacology experiments. We reported that plantar incision in mice induced anxiety-like behaviors and mechanical pain hypersensitivity, with a concurrent decrease of the oxytocin receptor (OTR) in the hippocampus. The immunofluorescence staining showed that the OTR were enriched in pyramidal neurons in CA3 subregion of hippocampus and which were highly co-expressed with the BK(Ca) channels in CA3 subregion. Intranasal OXT significantly ameliorated this postsurgical pain and associated anxiety in a dose-dependent manner, while Intra-CA3 microinjection of OTR antagonist atosiban or the BK(Ca) channel blocker paxilline reduced the effect of OXT in incisional mice. Moreover, intra-CA3 microinjection of BK(Ca) channel opener NS1619 produced a similar effect on postsurgical pain and associated anxiety-like behaviors as those observed following intranasal OXT administration. Conversely, intra-CA3 microinjection of BK(Ca) channel blocker paxilline in normal mice was sufficient to evoke mechanical pain hypersensitivity. Taken together, our data suggested that intranasal OXT administration exerted analgesic and anxiolytic effects in incisional mice by opening BK(Ca) channels in the CA3 subregion of hippocampus.
Collapse
Affiliation(s)
- Yan-Bin Tang
- Department of Anesthesiology and Perioperative Medicine, Fuzong Clinical Medical College (900th Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, PR China; Department of Anesthesiology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, PR China
| | - Li Tang
- Department of Orthodontics, The Affiliated Hospital of Qingdao University, School of Stomatology, Qingdao University, Qingdao, Shandong, PR China
| | - Bin Chen
- Department of Anesthesiology and Perioperative Medicine, Fuzong Clinical Medical College (900th Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, PR China
| | - Miao-Jie Fan
- Department of Anesthesiology and Perioperative Medicine, Fuzong Clinical Medical College (900th Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, PR China
| | - Gao-Jie Chen
- Department of Anesthesiology and Perioperative Medicine, Fuzong Clinical Medical College (900th Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, PR China
| | - Yu-Ning Ou
- Department of Anesthesiology and Perioperative Medicine, Fuzong Clinical Medical College (900th Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, PR China
| | - Fei Yang
- Department of Anesthesiology and Perioperative Medicine, Fuzong Clinical Medical College (900th Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, PR China.
| | - Xiao-Zhi Wu
- Department of Anesthesiology and Perioperative Medicine, Fuzong Clinical Medical College (900th Hospital of the Joint Logistic Support Force), Fujian Medical University, Fuzhou, Fujian, PR China
| |
Collapse
|
5
|
Park YS, Sung KW, Kim IB. IK Channel Confers Fine-tuning of Rod Bipolar Cell Excitation and Synaptic Transmission in the Retina. FUNCTION 2025; 6:zqae054. [PMID: 39716393 PMCID: PMC11815585 DOI: 10.1093/function/zqae054] [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: 07/26/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/25/2024] Open
Abstract
During retinal visual processing, rod bipolar cells (RBC) transfer scotopic signals from rods to AII amacrine cells as second-order neurons. Elucidation of the RBC's excitation/inhibition is essential for understanding the visual signal transmission. Excitation mechanisms via mGluR6 and voltage-gated Ca2+ channels in the RBCs and GABAergic inhibitory synaptic inputs have been studied in previous studies. However, its intrinsic inhibitory mechanisms like K+ and Cl- channels remain unclear. We focused on RBC's prominent K+ current, which exhibits voltage and Ca2+ dependence. We isolated and confirmed the expression of intermediate-conductance Ca2+-activated K+ channels (IK) in RBCs using the patch-clamp method with IK inhibitors (clotrimazole and TRAM34) and immunohistochemistry. The regulation of the IK channel primarily relies on Ca2+ influx via low-threshold Ca2+ channels during RBC's excitation. Additionally, IK mediates late repolarization and suppresses excessive oscillation of the membrane potential in the RBCs, enabling fast and transient synaptic transmission to AII amacrine cells. Our findings highlight the unique role of the IK channel in RBCs, suggesting that it plays a critical role in the scotopic pathway by fine-tuning RBC activity.
Collapse
Affiliation(s)
- Yong Soo Park
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Ki-Wug Sung
- Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - In-Beom Kim
- Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Catholic Institute for Applied Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
6
|
Martínez-Lazaro R, Reyes-Carrión A, Bartolomé-Martín D, Giraldez T. The NMDAR-BK channelosomes as regulators of synaptic plasticity. Biochem Soc Trans 2025; 53:BST20240425. [PMID: 39874044 DOI: 10.1042/bst20240425] [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: 09/29/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 01/30/2025]
Abstract
Large conductance voltage- and calcium-activated potassium channels (BK channels) are extensively found throughout the central nervous system and play a crucial role in various neuronal functions. These channels are activated by a combination of cell membrane depolarisation and an increase in intracellular calcium concentration, provided by calcium sources located close to BK. In 2001, Isaacson and Murphy first demonstrated the coupling of BK channels with N-methyl-D-aspartate receptors (NMDAR) in olfactory bulb neurons. Since then, additional evidence has confirmed this functional coupling in other brain regions and highlighted its significance in neuronal function and pathophysiology. In this review, we explore the current understanding of these macrocomplexes in the brain, the molecular mechanisms behind their interactions and their potential roles in neurodevelopmental disorders, paving the way for new treatment strategies.
Collapse
Affiliation(s)
- Rebeca Martínez-Lazaro
- Departamento de Ciencias Médicas Básicas, Facultad de Ciencias de la Salud-sección Medicina, Universidad de La Laguna, Tenerife, ES-38071, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Tenerife ES-38071, Spain
| | - Andrea Reyes-Carrión
- Departamento de Ciencias Médicas Básicas, Facultad de Ciencias de la Salud-sección Medicina, Universidad de La Laguna, Tenerife, ES-38071, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Tenerife ES-38071, Spain
| | - David Bartolomé-Martín
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Tenerife ES-38071, Spain
- Departamento de Bioquímica, Microbiología, Biología Celular y Genética, Facultad de Ciencias, Universidad de La Laguna, Tenerife, ES-38071, Spain
| | - Teresa Giraldez
- Departamento de Ciencias Médicas Básicas, Facultad de Ciencias de la Salud-sección Medicina, Universidad de La Laguna, Tenerife, ES-38071, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Tenerife ES-38071, Spain
| |
Collapse
|
7
|
Pal Mahadevan V, Galagovsky D, Knaden M, Hansson BS. Preference for and resistance to a toxic sulfur volatile opens up a unique niche in Drosophila busckii. Nat Commun 2025; 16:767. [PMID: 39824833 PMCID: PMC11742422 DOI: 10.1038/s41467-025-55971-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
The ability to tolerate otherwise toxic compounds can open up unique niches in nature. Among drosophilid flies, few examples of such adaptations are known and those which are known are typically from highly host-specific species. Here we show that the human commensal species Drosophila busckii uses dimethyldisulfide (DMDS) as a key mediator in its host selection. Despite DMDS's neurotoxic properties, D. busckii has evolved tolerance towards high concentrations and uses the compound as an olfactory cue to pinpoint food and oviposition sites. This adaptability is likely linked to insensitivity of the enzyme complex cytochrome c oxidase (COX), which is a DMDS target in other insects. Our findings position D. busckii as a potential model for studying resistance to toxic gases affecting COX and offers insight into evolutionary adaptations within specific ecological contexts.
Collapse
Affiliation(s)
- Venkatesh Pal Mahadevan
- Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Jena, Germany
- Max Planck Center next Generation Insect Chemical Ecology, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Diego Galagovsky
- Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Markus Knaden
- Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Jena, Germany
- Max Planck Center next Generation Insect Chemical Ecology, Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Bill S Hansson
- Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, Jena, Germany.
- Max Planck Center next Generation Insect Chemical Ecology, Max Planck Institute for Chemical Ecology, Jena, Germany.
| |
Collapse
|
8
|
Qiu J, Voliotis M, Bosch MA, Li XF, Zweifel LS, Tsaneva-Atanasova K, O'Byrne KT, Rønnekleiv OK, Kelly MJ. Estradiol elicits distinct firing patterns in arcuate nucleus kisspeptin neurons of females through altering ion channel conductances. eLife 2024; 13:RP96691. [PMID: 39671233 PMCID: PMC11643640 DOI: 10.7554/elife.96691] [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] [Indexed: 12/14/2024] Open
Abstract
Hypothalamic kisspeptin (Kiss1) neurons are vital for pubertal development and reproduction. Arcuate nucleus Kiss1 (Kiss1ARH) neurons are responsible for the pulsatile release of gonadotropin-releasing hormone (GnRH). In females, the behavior of Kiss1ARH neurons, expressing Kiss1, neurokinin B (NKB), and dynorphin (Dyn), varies throughout the ovarian cycle. Studies indicate that 17β-estradiol (E2) reduces peptide expression but increases Slc17a6 (Vglut2) mRNA and glutamate neurotransmission in these neurons, suggesting a shift from peptidergic to glutamatergic signaling. To investigate this shift, we combined transcriptomics, electrophysiology, and mathematical modeling. Our results demonstrate that E2 treatment upregulates the mRNA expression of voltage-activated calcium channels, elevating the whole-cell calcium current that contributes to high-frequency burst firing. Additionally, E2 treatment decreased the mRNA levels of canonical transient receptor potential (TPRC) 5 and G protein-coupled K+ (GIRK) channels. When Trpc5 channels in Kiss1ARH neurons were deleted using CRISPR/SaCas9, the slow excitatory postsynaptic potential was eliminated. Our data enabled us to formulate a biophysically realistic mathematical model of Kiss1ARH neurons, suggesting that E2 modifies ionic conductances in these neurons, enabling the transition from high-frequency synchronous firing through NKB-driven activation of TRPC5 channels to a short bursting mode facilitating glutamate release. In a low E2 milieu, synchronous firing of Kiss1ARH neurons drives pulsatile release of GnRH, while the transition to burst firing with high, preovulatory levels of E2 would facilitate the GnRH surge through its glutamatergic synaptic connection to preoptic Kiss1 neurons.
Collapse
Affiliation(s)
- Jian Qiu
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science UniversityPortlandUnited States
| | - Margaritis Voliotis
- Department of Mathematics and Statistics, University of ExeterExeterUnited Kingdom
- Living Systems Institute, University of ExeterExeterUnited Kingdom
| | - Martha A Bosch
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science UniversityPortlandUnited States
| | - Xiao Feng Li
- Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College LondonLondonUnited Kingdom
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of WashingtonSeattleUnited States
- Depatment of Pharmacology, University of WashingtonSeattleUnited States
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics and Statistics, University of ExeterExeterUnited Kingdom
- Living Systems Institute, University of ExeterExeterUnited Kingdom
| | - Kevin T O'Byrne
- Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College LondonLondonUnited Kingdom
| | - Oline K Rønnekleiv
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science UniversityPortlandUnited States
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Martin J Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science UniversityPortlandUnited States
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| |
Collapse
|
9
|
Ferraguto C, Piquemal-Lagoueillat M, Lemaire V, Moreau MM, Trazzi S, Uguagliati B, Ciani E, Bertrand SS, Louette E, Bontempi B, Pietropaolo S. Therapeutic efficacy of the BKCa channel opener chlorzoxazone in a mouse model of Fragile X syndrome. Neuropsychopharmacology 2024; 49:2032-2041. [PMID: 39223257 PMCID: PMC11480417 DOI: 10.1038/s41386-024-01956-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/30/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Fragile X syndrome (FXS) is an X-linked neurodevelopmental disorder characterized by several behavioral abnormalities, including hyperactivity, anxiety, sensory hyper-responsiveness, and autistic-like symptoms such as social deficits. Despite considerable efforts, effective pharmacological treatments are still lacking, prompting the need for exploring the therapeutic value of existing drugs beyond their original approved use. One such repurposed drug is chlorzoxazone which is classified as a large-conductance calcium-dependent potassium (BKCa) channel opener. Reduced BKCa channel functionality has been reported in FXS patients, suggesting that molecules activating these channels could serve as promising treatments for this syndrome. Here, we sought to characterize the therapeutic potential of chlorzoxazone using the Fmr1-KO mouse model of FXS which recapitulates the main phenotypes of FXS, including BKCa channel alterations. Chlorzoxazone, administered either acutely or chronically, rescued hyperactivity and acoustic hyper-responsiveness as well as impaired social interactions exhibited by Fmr1-KO mice. Chlorzoxazone was more efficacious in alleviating these phenotypes than gaboxadol and metformin, two repurposed treatments for FXS that do not target BKCa channels. Systemic administration of chlorzoxazone modulated the neuronal activity-dependent gene c-fos in selected brain areas of Fmr1-KO mice, corrected aberrant hippocampal dendritic spines, and was able to rescue impaired BKCa currents recorded from hippocampal and cortical neurons of these mutants. Collectively, these findings provide further preclinical support for BKCa channels as a valuable therapeutic target for treating FXS and encourage the repurposing of chlorzoxazone for clinical applications in FXS and other related neurodevelopmental diseases.
Collapse
Affiliation(s)
| | | | - Valerie Lemaire
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, Bordeaux, France
| | - Maïté M Moreau
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | - Stefania Trazzi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Beatrice Uguagliati
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Elisabetta Ciani
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | | | | | - Bruno Bontempi
- Univ. Bordeaux, CNRS, EPHE, INCIA, UMR 5287, Bordeaux, France
| | | |
Collapse
|
10
|
Qiu J, Voliotis M, Bosch MA, Li XF, Zweifel LS, Tsaneva-Atanasova K, O’Byrne KT, Rønnekleiv OK, Kelly MJ. Estradiol elicits distinct firing patterns in arcuate nucleus kisspeptin neurons of females through altering ion channel conductances. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.20.581121. [PMID: 38915596 PMCID: PMC11195100 DOI: 10.1101/2024.02.20.581121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Hypothalamic kisspeptin (Kiss1) neurons are vital for pubertal development and reproduction. Arcuate nucleus Kiss1 (Kiss1ARH) neurons are responsible for the pulsatile release of Gonadotropin-releasing Hormone (GnRH). In females, the behavior of Kiss1ARH neurons, expressing Kiss1, Neurokinin B (NKB), and Dynorphin (Dyn), varies throughout the ovarian cycle. Studies indicate that 17β-estradiol (E2) reduces peptide expression but increases Vglut2 mRNA and glutamate neurotransmission in these neurons, suggesting a shift from peptidergic to glutamatergic signaling. To investigate this shift, we combined transcriptomics, electrophysiology, and mathematical modeling. Our results demonstrate that E2 treatment upregulates the mRNA expression of voltage-activated calcium channels, elevating the whole-cell calcium current and that contribute to high-frequency burst firing. Additionally, E2 treatment decreased the mRNA levels of Canonical Transient Receptor Potential (TPRC) 5 and G protein-coupled K+ (GIRK) channels. When TRPC5 channels in Kiss1ARH neurons were deleted using CRISPR, the slow excitatory postsynaptic potential (sEPSP) was eliminated. Our data enabled us to formulate a biophysically realistic mathematical model of the Kiss1ARH neuron, suggesting that E2 modifies ionic conductances in Kiss1ARH neurons, enabling the transition from high frequency synchronous firing through NKB-driven activation of TRPC5 channels to a short bursting mode facilitating glutamate release. In a low E2 milieu, synchronous firing of Kiss1ARH neurons drives pulsatile release of GnRH, while the transition to burst firing with high, preovulatory levels of E2 would facilitate the GnRH surge through its glutamatergic synaptic connection to preoptic Kiss1 neurons.
Collapse
Affiliation(s)
- Jian Qiu
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science U., Portland, OR 97239, USA
| | - Margaritis Voliotis
- Department of Mathematics and Statistics, University of Exeter, Stocker Rd, Exeter, EX4 4PY, UK
- Living Systems Institute, University of Exeter, Stocker Rd, Exeter, EX4 4PY, UK
| | - Martha A. Bosch
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science U., Portland, OR 97239, USA
| | - Xiao Feng Li
- Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London, Guy’s Campus, London SE1 1UL, UK
| | - Larry S. Zweifel
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA
- Depatment of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics and Statistics, University of Exeter, Stocker Rd, Exeter, EX4 4PY, UK
- Living Systems Institute, University of Exeter, Stocker Rd, Exeter, EX4 4PY, UK
| | - Kevin T. O’Byrne
- Department of Women and Children’s Health, School of Life Course and Population Sciences, King’s College London, Guy’s Campus, London SE1 1UL, UK
| | - Oline K. Rønnekleiv
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science U., Portland, OR 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Martin J. Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science U., Portland, OR 97239, USA
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| |
Collapse
|
11
|
Kshatri A, Rivero-Pérez B, Giraldez T. Subunit-specific inhibition of BK channels by piperine. Biophys J 2024; 123:1942-1953. [PMID: 37700524 PMCID: PMC11309970 DOI: 10.1016/j.bpj.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 08/03/2023] [Accepted: 09/08/2023] [Indexed: 09/14/2023] Open
Abstract
Piperine is the principal alkaloid present in black pepper and is well-known for its diverse pharmacological effects, including inhibition of different ion channels. Large conductance Ca2+-activated K+ channels (BK) are widely expressed across several tissues and play a vital role in many physiological functions. In this study, we investigated the pharmacological effects of piperine on various BK channel subunit compositions (BKα, BKαβ1,4, BKαγ1,3) expressed in HEK293T cells. Piperine in zero Ca2+ reversibly inhibited currents from the pore-forming BKα channels in a dose-dependent manner with a half-maximal inhibitory concentration (IC50) of 4.8 μM. Elevating the internal Ca2+ concentration from 0 to 100 μM significantly attenuated the inhibitory effects of piperine on BKα channels. The mutation G311S in the pore domain failed to alter the modulatory effects of piperine, whereas deletion of the entire cytoplasmic domain from BKα channels ablated its inhibitory effects. Addition of either BKβ1 or β4 regulatory subunits did not alter the efficacy of piperine on BKα channels. Interestingly, co-expression of either BKγ1 or BKγ3 subunits greatly diminished the ability of piperine to inhibit BKα channels. Our findings demonstrate that piperine is a potent natural modulator of BKα/BKαβ1,4 subunits but not BKαγ1,3 subunits. The mechanism of piperine modulation appeared to be allosteric and differs from that of other BK pore blockers (paxilline, peptide toxins, and quaternary ammonium compounds). Together, our results unravel the potential of piperine to inhibit BK channels, providing a new tool to explore mechanisms underlying the effects of regulatory subunits.
Collapse
Affiliation(s)
- Aravind Kshatri
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Belinda Rivero-Pérez
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain
| | - Teresa Giraldez
- Department of Basic Medical Sciences, Medical School, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologias Biomedicas, Universidad de La Laguna, Tenerife, Spain.
| |
Collapse
|
12
|
Abdalla MMI. Insulin resistance as the molecular link between diabetes and Alzheimer's disease. World J Diabetes 2024; 15:1430-1447. [PMID: 39099819 PMCID: PMC11292327 DOI: 10.4239/wjd.v15.i7.1430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/08/2024] [Accepted: 05/06/2024] [Indexed: 07/08/2024] Open
Abstract
Diabetes mellitus (DM) and Alzheimer's disease (AD) are two major health concerns that have seen a rising prevalence worldwide. Recent studies have indicated a possible link between DM and an increased risk of developing AD. Insulin, while primarily known for its role in regulating blood sugar, also plays a vital role in protecting brain functions. Insulin resistance (IR), especially prevalent in type 2 diabetes, is believed to play a significant role in AD's development. When insulin signalling becomes dysfunctional, it can negatively affect various brain functions, making individuals more susceptible to AD's defining features, such as the buildup of beta-amyloid plaques and tau protein tangles. Emerging research suggests that addressing insulin-related issues might help reduce or even reverse the brain changes linked to AD. This review aims to explore the rela-tionship between DM and AD, with a focus on the role of IR. It also explores the molecular mechanisms by which IR might lead to brain changes and assesses current treatments that target IR. Understanding IR's role in the connection between DM and AD offers new possibilities for treatments and highlights the importance of continued research in this interdisciplinary field.
Collapse
Affiliation(s)
- Mona Mohamed Ibrahim Abdalla
- Department of Human Biology, School of Medicine, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| |
Collapse
|
13
|
Parikh A, Krogman W, Walker J. The impact of volatile anesthetics and propofol on phosphatidylinositol 4,5-bisphosphate signaling. Arch Biochem Biophys 2024; 757:110045. [PMID: 38801966 DOI: 10.1016/j.abb.2024.110045] [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: 02/14/2024] [Revised: 04/29/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Phosphatidylinositol 4,5-bisphosphate (PIP2), as well as other anionic phospholipids, play a pivotal role in various cellular processes, including ion channel regulation, receptor trafficking, and intracellular signaling pathways. The binding of volatile anesthetics and propofol to PIP2 leads to alterations in PIP2-mediated signaling causing modulation of ion channels such as ɣ-aminobutyric acid type A (GABAA) receptors, voltage-gated calcium channels, and potassium channels through various mechanisms. Additionally, the interaction between anionic phospholipids and G protein-coupled receptors plays a critical role in various anesthetic pathways, with these anesthetic-induced changes impacting PIP2 levels which cause cascading effects on receptor trafficking, including GABAA receptor internalization. This comprehensive review of various mechanisms of interaction provides insights into the intricate interplay between PIP2 signaling and anesthetic-induced changes, shedding light on the molecular mechanisms underlying anesthesia.
Collapse
Affiliation(s)
- Ayaan Parikh
- Wichita Collegiate School, Wichita, KS. 9115 E 13th St N, Wichita, KS, 67206, USA.
| | - William Krogman
- University of Kansas School of Medicine-Wichita, Wichita, KS, USA; Department of Anesthesiology, 929 N St Francis, Room 8079, Wichita, KS, 67214, USA
| | - James Walker
- University of Kansas School of Medicine-Wichita, Wichita, KS, USA; Department of Anesthesiology, 929 N St Francis, Room 8079, Wichita, KS, 67214, USA
| |
Collapse
|
14
|
Wang H, Zuo W, Feng X, Huo X, Liang Y, Wang B, Sharma D, Li X, Yasin B, Ye JH, Hu H, Tao YX. ESRRG-controlled downregulation of KCNN1 in primary sensory neurons is required for neuropathic pain. JCI Insight 2024; 9:e180085. [PMID: 38912580 PMCID: PMC11383585 DOI: 10.1172/jci.insight.180085] [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: 02/06/2024] [Accepted: 05/02/2024] [Indexed: 06/25/2024] Open
Abstract
Peripheral nerve injury-induced neuronal hyperactivity in the dorsal root ganglion (DRG) participates in neuropathic pain. The calcium-activated potassium channel subfamily N member 1 (KCNN1) mediates action potential afterhyperpolarization (AHP) and gates neuronal excitability. However, the specific contribution of DRG KCNN1 to neuropathic pain is not yet clear. We report that chronic constriction injury (CCI) of the unilateral sciatic nerve or unilateral ligation of the fourth lumbar nerve produced the downregulation of Kcnn1 mRNA and KCNN1 protein in the injured DRG. This downregulation was partially attributed to a decrease in DRG estrogen-related receptor gamma (ESRRG), a transcription factor, which led to reduced binding to the Kcnn1 promoter. Rescuing this downregulation prevented CCI-induced decreases in total potassium voltage currents and AHP currents, reduced excitability in the injured DRG neurons, and alleviated CCI-induced development and maintenance of nociceptive hypersensitivities, without affecting locomotor function and acute pain. Mimicking the CCI-induced DRG KCNN1 downregulation resulted in augmented responses to mechanical, heat, and cold stimuli in naive mice. Our findings indicate that ESRRG-controlled downregulation of DRG KCNN1 is likely essential for the development and maintenance of neuropathic pain. Thus, KCNN1 may serve as a potential target for managing this disorder.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jiang-Hong Ye
- Department of Anesthesiology
- Department of Physiology, Pharmacology & Neuroscience; and
| | - Huijuan Hu
- Department of Anesthesiology
- Department of Physiology, Pharmacology & Neuroscience; and
| | - Yuan-Xiang Tao
- Department of Anesthesiology
- Department of Physiology, Pharmacology & Neuroscience; and
- Department of Cell Biology & Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| |
Collapse
|
15
|
Segura É, Zhao J, Broszczak M, Audet F, Sauvé R, Parent L. Investigating the Impact of Electrostatic Interactions on Calmodulin Binding and Ca 2+-Dependent Activation of the Calcium-Gated Potassium SK4 Channel. Int J Mol Sci 2024; 25:4255. [PMID: 38673845 PMCID: PMC11050286 DOI: 10.3390/ijms25084255] [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: 03/08/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Ca2+ binding to the ubiquitous Ca2+ sensing protein calmodulin (CaM) activates the intermediate conductance Ca2+-activated SK4 channel. Potential hydrophilic pockets for CaM binding have been identified at the intracellular HA and HB helices in the C-terminal of SK4 from the three published cryo-EM structures of SK4. Single charge reversal substitutions at either site, significantly weakened the pull-down of SK4 by CaM wild-type (CaM), and decreased the TRAM-34 sensitive outward K+ current densities in native HEK293T cells when compared with SK4 WT measured under the same conditions. Only the doubly substituted SK4 R352D/R355D (HB helix) obliterated the CaM-mediated pull-down and thwarted outward K+ currents. However, overexpression of CaM E84K/E87K, which had been predicted to face the arginine doublet, restored the CaM-mediated pull-down of SK4 R352D/R355D and normalized its whole-cell current density. Virtual analysis of the putative salt bridges supports a unique role for the positively charged arginine doublet at the HB helix into anchoring the interaction with the negatively charged CaM glutamate 84 and 87 CaM. Our findings underscore the unique contribution of electrostatic interactions in carrying CaM binding onto SK4 and support the role of the C-terminal HB helix to the Ca2+-dependent gating process.
Collapse
Affiliation(s)
- Émilie Segura
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Centre de Recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, QC H1T 1C8, Canada; (É.S.); (F.A.)
| | - Juan Zhao
- Centre de Recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, QC H1T 1C8, Canada; (J.Z.); (M.B.)
| | - Marlena Broszczak
- Centre de Recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, QC H1T 1C8, Canada; (J.Z.); (M.B.)
| | - Frédéric Audet
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Centre de Recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, QC H1T 1C8, Canada; (É.S.); (F.A.)
| | - Rémy Sauvé
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, 2900 Bd Édouard-Montpetit, Montréal, QC H3T 1J4, Canada;
| | - Lucie Parent
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Centre de Recherche de l’Institut de Cardiologie de Montréal, Université de Montréal, Montréal, QC H1T 1C8, Canada; (É.S.); (F.A.)
| |
Collapse
|
16
|
Maiorov SA, Kairat BK, Berezhnov AV, Zinchenko VP, Gaidin SG, Kosenkov AM. Peculiarities of ion homeostasis in neurons containing calcium-permeable AMPA receptors. Arch Biochem Biophys 2024; 754:109951. [PMID: 38452968 DOI: 10.1016/j.abb.2024.109951] [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: 09/22/2023] [Revised: 02/16/2024] [Accepted: 02/28/2024] [Indexed: 03/09/2024]
Abstract
Glutamate excitotoxicity accompanies numerous brain pathologies, including traumatic brain injury, ischemic stroke, and epilepsy. Disturbances of the ion homeostasis, mitochondria dysfunction, and further cell death are considered the main detrimental consequences of excitotoxicity. It is well known that neurons demonstrate different vulnerability to pathological exposures. In this regard, neurons containing calcium-permeable AMPA receptors (CP-AMPARs) may show higher susceptibility to excitotoxicity due to an additional pathway of Ca2+ influx. Here, we demonstrate that neurons containing CP-AMPARs are characterized by the higher amplitude of the glutamate-induced elevation of intracellular Ca2+ concentration ([Ca2+]i) and slower restoration of [Ca2+]i level compared to non-CP-AMPA neurons. Moreover, we have found that NASPM, an antagonist of CP-AMPARs, significantly decreases the amplitude of the [Ca2+]i elevation induced by glutamate or selective AMPARs agonist, 5-fluorowillardiine. In contrast, the antagonists of NMDARs or KARs affect insignificantly. We have also described some peculiarities of Na+, K+, and H+ intracellular dynamics in neurons containing CP-AMPARs. In particular, the amplitude of [Na+]i elevation was lower compared to non-CP-AMPA neurons, whereas the amplitude of [K+]i decrease was higher. We have shown the significant inverse correlation between [K+]i and [Ca2+]i and between intracellular pH and [Na+]i in CP-AMPARs-containing and non-CP-AMPA neurons upon glutamate excitotoxicity. Our data indicate that CP-AMPARs-mediated Ca2+ influx and slow removal of Ca2+ from the cytosol may underlie the vulnerability of the CP-AMPARs-containing neurons to glutamate excitotoxicity. Further studies of the mechanisms mediating the disturbances in ion homeostasis are crucial for developing new approaches for protecting these neurons at brain pathologies.
Collapse
Affiliation(s)
- Sergei A Maiorov
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290, Pushchino, Russia
| | | | - Alexey V Berezhnov
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290, Pushchino, Russia
| | - Valery P Zinchenko
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290, Pushchino, Russia
| | - Sergei G Gaidin
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290, Pushchino, Russia.
| | - Artem M Kosenkov
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290, Pushchino, Russia.
| |
Collapse
|
17
|
Yang H, Zhang X, Zhang M, Lu Y, Xie B, Sun S, Yu H, Cong B, Luo Y, Ma C, Wen D. Roles of lncLingo2 and its derived miR-876-5p in the acquisition of opioid reinforcement. Addict Biol 2024; 29:e13375. [PMID: 38380802 PMCID: PMC10898844 DOI: 10.1111/adb.13375] [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: 11/14/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 02/22/2024]
Abstract
Recent studies found that non-coding RNAs (ncRNAs) played crucial roles in drug addiction through epigenetic regulation of gene expression and underlying drug-induced neuroadaptations. In this study, we characterized lncRNA transcriptome profiles in the nucleus accumbens (NAc) of mice exhibiting morphine-conditioned place preference (CPP) and explored the prospective roles of novel differentially expressed lncRNA, lncLingo2 and its derived miR-876-5p in the acquisition of opioids-associated behaviours. We found that the lncLingo2 was downregulated within the NAc core (NAcC) but not in the NAc shell (NAcS). This downregulation was found to be associated with the development of morphine CPP and heroin intravenous self-administration (IVSA). As Mfold software revealed that the secondary structures of lncLingo2 contained the sequence of pre-miR-876, transfection of LV-lncLingo2 into HEK293 cells significantly upregulated miR-876 expression and the changes of mature miR-876 are positively correlated with lncLingo2 expression in NAcC of morphine CPP trained mice. Delivering miR-876-5p mimics into NAcC also inhibited the acquisition of morphine CPP. Furthermore, bioinformatics analysis and dual-luciferase assay confirmed that miR-876-5p binds to its target gene, Kcnn3, selectively and regulates morphine CPP training-induced alteration of Kcnn3 expression. Lastly, the electrophysiological analysis indicated that the currents of small conductance calcium-activated potassium (SK) channel was increased, which led to low neuronal excitability in NAcC after CPP training, and these changes were reversed by lncLingo2 overexpression. Collectively, lncLingo2 may function as a precursor of miR-876-5p in NAcC, hence modulating the development of opioid-associated behaviours in mice, which may serve as an underlying biomarker and therapeutic target of opioid addiction.
Collapse
Affiliation(s)
- Hongyu Yang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Xiuning Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Minglong Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
- Department of GeneticsQiqihar Medical UniversityQiqiharHeilongjiang ProvinceChina
| | - Yun Lu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Shaoguang Sun
- Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei ProvinceHebei Medical UniversityShijiazhuangChina
- Key Laboratory of Neural and Vascular BiologyMinistry of EducationShijiazhuangHebei ProvinceChina
| | - Hailei Yu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
| | - Yixiao Luo
- Hunan Province People's HospitalThe First‐Affiliated Hospital of Hunan Normal UniversityChangshaChina
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
- Key Laboratory of Neural and Vascular BiologyMinistry of EducationShijiazhuangHebei ProvinceChina
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and ToxicologyChinese Academy of Medical SciencesShijiazhuangHebei ProvinceChina
- Key Laboratory of Neural and Vascular BiologyMinistry of EducationShijiazhuangHebei ProvinceChina
| |
Collapse
|
18
|
Liu Y, Xia D, Zhong L, Chen L, Zhang L, Ai M, Mei R, Pang R. Casein Kinase 2 Affects Epilepsy by Regulating Ion Channels: A Potential Mechanism. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:894-905. [PMID: 37350003 DOI: 10.2174/1871527322666230622124618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 03/31/2023] [Accepted: 04/10/2023] [Indexed: 06/24/2023]
Abstract
Epilepsy, characterized by recurrent seizures and abnormal brain discharges, is the third most common chronic disorder of the Central Nervous System (CNS). Although significant progress has been made in the research on antiepileptic drugs (AEDs), approximately one-third of patients with epilepsy are refractory to these drugs. Thus, research on the pathogenesis of epilepsy is ongoing to find more effective treatments. Many pathological mechanisms are involved in epilepsy, including neuronal apoptosis, mossy fiber sprouting, neuroinflammation, and dysfunction of neuronal ion channels, leading to abnormal neuronal excitatory networks in the brain. CK2 (Casein kinase 2), which plays a critical role in modulating neuronal excitability and synaptic transmission, has been shown to be associated with epilepsy. However, there is limited research on the mechanisms involved. Recent studies have suggested that CK2 is involved in regulating the function of neuronal ion channels by directly phosphorylating them or their binding partners. Therefore, in this review, we will summarize recent research advances regarding the potential role of CK2 regulating ion channels in epilepsy, aiming to provide more evidence for future studies.
Collapse
Affiliation(s)
- Yan Liu
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Di Xia
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Lianmei Zhong
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Ling Chen
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
- Yunnan Provincial Clinical Research Center for Neurological Disease, Kunming, Yunnan, 650032, China
| | - Linming Zhang
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Mingda Ai
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Rong Mei
- Department of Neurology, the First People's Hospital of Yunnan Province, Kunming, Yunnan, 650034, China
| | - Ruijing Pang
- Department of Neurology, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| |
Collapse
|
19
|
Silván Á, Kohlmeier KA, Herrik KF, Hougaard C. Gating small conductance calcium-activated potassium channels in the thalamic reticular nucleus. Synapse 2024; 78:e22283. [PMID: 37837643 DOI: 10.1002/syn.22283] [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: 04/14/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/16/2023]
Abstract
Small conductance calcium-activated potassium (SK) channels are well-known regulators of neuronal excitability. In the thalamic hub, SK2 channels act as pacemakers of thalamic reticular neurons, which play a key role in the thalamocortical circuit. Several disease-linked genes are highly enriched in these neurons, including genes known to be associated with schizophrenia and attentional disorders, which could affect neuronal firing. The present study assessed the effect of pharmacological modulation of SK channels in the firing pattern and intrinsic properties of thalamic reticular neurons by performing whole cell patch clamp recordings in brain slices. Two SK positive allosteric modulators and one negative allosteric modulator were used: CyPPA, NS309, and NS8593, respectively. By acting on the burst afterhyperpolarization (AHP), negative modulation of SK channels resulted in increased action potential (AP) firing, increased burst duration, and decreased intervals between bursts. Conversely, both CyPPA and NS309 increased the afterburst AHP, prolonging the interburst interval, which additionally resulted in reduced AP firing in the case of NS309. Alterations in SK channel activity would be expected to alter functioning of thalamocortical circuits. Targeting SK channels could be promising in treating disorders involving thalamic reticular dysfunction such as psychiatric and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ágata Silván
- H Lundbeck A/S, Circuit Biology, Valby, Denmark
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Kristi Anne Kohlmeier
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
20
|
Alexander SPH, Mathie AA, Peters JA, Veale EL, Striessnig J, Kelly E, Armstrong JF, Faccenda E, Harding SD, Davies JA, Aldrich RW, Attali B, Baggetta AM, Becirovic E, Biel M, Bill RM, Caceres AI, Catterall WA, Conner AC, Davies P, De Clerq K, Delling M, Di Virgilio F, Falzoni S, Fenske S, Fortuny-Gomez A, Fountain S, George C, Goldstein SAN, Grimm C, Grissmer S, Ha K, Hammelmann V, Hanukoglu I, Hu M, Ijzerman AP, Jabba SV, Jarvis M, Jensen AA, Jordt SE, Kaczmarek LK, Kellenberger S, Kennedy C, King B, Kitchen P, Liu Q, Lynch JW, Meades J, Mehlfeld V, Nicke A, Offermanns S, Perez-Reyes E, Plant LD, Rash L, Ren D, Salman MM, Sieghart W, Sivilotti LG, Smart TG, Snutch TP, Tian J, Trimmer JS, Van den Eynde C, Vriens J, Wei AD, Winn BT, Wulff H, Xu H, Yang F, Fang W, Yue L, Zhang X, Zhu M. The Concise Guide to PHARMACOLOGY 2023/24: Ion channels. Br J Pharmacol 2023; 180 Suppl 2:S145-S222. [PMID: 38123150 PMCID: PMC11339754 DOI: 10.1111/bph.16178] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
The Concise Guide to PHARMACOLOGY 2023/24 is the sixth in this series of biennial publications. The Concise Guide provides concise overviews, mostly in tabular format, of the key properties of approximately 1800 drug targets, and over 6000 interactions with about 3900 ligands. There is an emphasis on selective pharmacology (where available), plus links to the open access knowledgebase source of drug targets and their ligands (https://www.guidetopharmacology.org/), which provides more detailed views of target and ligand properties. Although the Concise Guide constitutes almost 500 pages, the material presented is substantially reduced compared to information and links presented on the website. It provides a permanent, citable, point-in-time record that will survive database updates. The full contents of this section can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.16178. Ion channels are one of the six major pharmacological targets into which the Guide is divided, with the others being: G protein-coupled receptors, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. The landscape format of the Concise Guide is designed to facilitate comparison of related targets from material contemporary to mid-2023, and supersedes data presented in the 2021/22, 2019/20, 2017/18, 2015/16 and 2013/14 Concise Guides and previous Guides to Receptors and Channels. It is produced in close conjunction with the Nomenclature and Standards Committee of the International Union of Basic and Clinical Pharmacology (NC-IUPHAR), therefore, providing official IUPHAR classification and nomenclature for human drug targets, where appropriate.
Collapse
Affiliation(s)
- Stephen P H Alexander
- School of Life Sciences, University of Nottingham Medical School, Nottingham, NG7 2UH, UK
| | - Alistair A Mathie
- School of Engineering, Arts, Science and Technology, University of Suffolk, Ipswich, IP4 1QJ, UK
| | - John A Peters
- Neurosci-ence Division, Medical Education Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Emma L Veale
- Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Anson Building, Central Avenue, Chatham Maritime, Chatham, Kent, ME4 4TB, UK
| | - Jörg Striessnig
- Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck, A-6020, Innsbruck, Austria
| | - Eamonn Kelly
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK
| | - Jane F Armstrong
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Elena Faccenda
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Simon D Harding
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Jamie A Davies
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | | | | | | | | | - Martin Biel
- Ludwig Maximilian University of Munich, Munich, Germany
| | | | | | | | | | - Paul Davies
- Tufts University School of Medicine, Boston, USA
| | | | - Markus Delling
- University of California San Francisco, San Francisco, USA
| | | | | | | | | | | | - Chandy George
- Nanyang Technological University, Singapore, Singapore
| | | | | | | | - Kotdaji Ha
- University of California San Francisco, San Francisco, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Annette Nicke
- Ludwig Maximilian University of Munich, Munich, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research/JW Goethe University, Bad Nauheim/Frankfurt, Germany
| | | | | | | | - Dejian Ren
- University of Pennsylvania, Philadelphia, USA
| | | | | | | | | | | | - Jinbin Tian
- University of Texas at Houston, Houston, USA
| | | | | | | | | | | | | | | | | | | | - Lixia Yue
- University of Connecticut, Farmington, USA
| | | | - Michael Zhu
- University of Texas at Houston, Houston, USA
| |
Collapse
|
21
|
Figueiredo IAD, Ferreira SRD, Fernandes JM, Silva BA, Vasconcelos LHC, Cavalcante FA. A review of the pathophysiology and the role of ion channels on bronchial asthma. Front Pharmacol 2023; 14:1236550. [PMID: 37841931 PMCID: PMC10568497 DOI: 10.3389/fphar.2023.1236550] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/08/2023] [Indexed: 10/17/2023] Open
Abstract
Asthma is one of the main non-communicable chronic diseases and affects a huge portion of the population. It is a multifactorial disease, classified into several phenotypes, being the allergic the most frequent. The pathophysiological mechanism of asthma involves a Th2-type immune response, with high concentrations of allergen-specific immunoglobulin E, eosinophilia, hyperreactivity and airway remodeling. These mechanisms are orchestrated by intracellular signaling from effector cells, such as lymphocytes and eosinophils. Ion channels play a fundamental role in maintaining the inflammatory response on asthma. In particular, transient receptor potential (TRP), stock-operated Ca2+ channels (SOCs), Ca2+-activated K+ channels (IKCa and BKCa), calcium-activated chloride channel (TMEM16A), cystic fibrosis transmembrane conductance regulator (CFTR), piezo-type mechanosensitive ion channel component 1 (PIEZO1) and purinergic P2X receptor (P2X). The recognition of the participation of these channels in the pathological process of asthma is important, as they become pharmacological targets for the discovery of new drugs and/or pharmacological tools that effectively help the pharmacotherapeutic follow-up of this disease, as well as the more specific mechanisms involved in worsening asthma.
Collapse
Affiliation(s)
- Indyra Alencar Duarte Figueiredo
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Sarah Rebeca Dantas Ferreira
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Jayne Muniz Fernandes
- Graduação em Farmácia, Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Bagnólia Araújo da Silva
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Luiz Henrique César Vasconcelos
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
- Departamento de Fisiologia e Patologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| | - Fabiana de Andrade Cavalcante
- Programa de Pós-graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
- Departamento de Fisiologia e Patologia, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa, Paraíba, Brazil
| |
Collapse
|
22
|
Wang B, Ma L, Guo X, Du S, Feng X, Liang Y, Govindarajalu G, Wu S, Liu T, Li H, Patel S, Bekker A, Hu H, Tao YX. A sensory neuron-specific long non-coding RNA reduces neuropathic pain by rescuing KCNN1 expression. Brain 2023; 146:3866-3884. [PMID: 37012681 PMCID: PMC10473565 DOI: 10.1093/brain/awad110] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 02/21/2023] [Accepted: 03/10/2023] [Indexed: 04/05/2023] Open
Abstract
Nerve injury to peripheral somatosensory system causes refractory neuropathic pain. Maladaptive changes of gene expression in primary sensory neurons are considered molecular basis of this disorder. Long non-coding RNAs (lncRNAs) are key regulators of gene transcription; however, their significance in neuropathic pain remains largely elusive.Here, we reported a novel lncRNA, named sensory neuron-specific lncRNA (SS-lncRNA), for its expression exclusively in dorsal root ganglion (DRG) and trigeminal ganglion. SS-lncRNA was predominantly expressed in small DRG neurons and significantly downregulated due to a reduction of early B cell transcription factor 1 in injured DRG after nerve injury. Rescuing this downregulation reversed a decrease of the calcium-activated potassium channel subfamily N member 1 (KCNN1) in injured DRG and alleviated nerve injury-induced nociceptive hypersensitivity. Conversely, DRG downregulation of SS-lncRNA reduced the expression of KCNN1, decreased total potassium currents and afterhyperpolarization currents and increased excitability in DRG neurons and produced neuropathic pain symptoms.Mechanistically, downregulated SS-lncRNA resulted in the reductions of its binding to Kcnn1 promoter and heterogeneous nuclear ribonucleoprotein M (hnRNPM), consequent recruitment of less hnRNPM to the Kcnn1 promoter and silence of Kcnn1 gene transcription in injured DRG.These findings indicate that SS-lncRNA may relieve neuropathic pain through hnRNPM-mediated KCNN1 rescue in injured DRG and offer a novel therapeutic strategy specific for this disorder.
Collapse
Affiliation(s)
- Bing Wang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Longfei Ma
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Xinying Guo
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Shibin Du
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Xiaozhou Feng
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Yingping Liang
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Gokulapriya Govindarajalu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Shaogen Wu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Tong Liu
- Center for Advanced Proteomics Research, Departments of Biochemistry, Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Hong Li
- Center for Advanced Proteomics Research, Departments of Biochemistry, Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Shivam Patel
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Alex Bekker
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Huijuan Hu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
- Department of Physiology, Pharmacology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Yuan-Xiang Tao
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
- Department of Physiology, Pharmacology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
23
|
Paul A, Chumbale SS, Lakra A, Kumar V, Alhat DS, Singh S. Insights into Leishmania donovani potassium channel family and their biological functions. 3 Biotech 2023; 13:266. [PMID: 37425093 PMCID: PMC10326225 DOI: 10.1007/s13205-023-03692-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 06/26/2023] [Indexed: 07/11/2023] Open
Abstract
Leishmania donovani is the causative organism for visceral leishmaniasis. Although this parasite was discovered over a century ago, nothing is known about role of potassium channels in L. donovani. Potassium channels are known for their crucial roles in cellular functions in other organisms. Recently the presence of a calcium-activated potassium channel in L. donovani was reported which prompted us to look for other proteins which could be potassium channels and to investigate their possible physiological roles. Twenty sequences were identified in L. donovani genome and subjected to estimation of physio-chemical properties, motif analysis, localization prediction and transmembrane domain analysis. Structural predictions were also done. The channels were majorly α-helical and predominantly localized in cell membrane and lysosomes. The signature selectivity filter of potassium channel was present in all the sequences. In addition to the conventional potassium channel activity, they were associated with gene ontology terms for mitotic cell cycle, cell death, modulation by virus of host process, cell motility etc. The entire study indicates the presence of potassium channel families in L. donovani which may have involvement in several cellular pathways. Further investigations on these putative potassium channels are needed to elucidate their roles in Leishmania. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-023-03692-y.
Collapse
Affiliation(s)
- Anindita Paul
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062 Punjab India
| | - Shubham Sunil Chumbale
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062 Punjab India
| | - Anjana Lakra
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062 Punjab India
| | - Vijay Kumar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062 Punjab India
| | - Dhanashri Sudam Alhat
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062 Punjab India
| | - Sushma Singh
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, 160062 Punjab India
| |
Collapse
|
24
|
Zhang M, Luo Y, Wang J, Sun Y, Xie B, Zhang L, Cong B, Ma C, Wen D. Roles of nucleus accumbens shell small-conductance calcium-activated potassium channels in the conditioned fear freezing. J Psychiatr Res 2023; 163:180-194. [PMID: 37216772 DOI: 10.1016/j.jpsychires.2023.05.057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/27/2023] [Accepted: 05/15/2023] [Indexed: 05/24/2023]
Abstract
BACKGROUND Posttraumatic stress disorder (PTSD), a psychiatric disorder caused by stressful events, is characterized by long-lasting fear memory. The nucleus accumbens shell (NAcS) is a key brain region that regulates fear-associated behavior. Small-conductance calcium-activated potassium channels (SK channels) play a key role in regulating the excitability of NAcS medium spiny neurons (MSNs) but their mechanisms of action in fear freezing are unclear. METHOD We established an animal model of traumatic memory using conditioned fear freezing paradigm, and investigated the alterations in SK channels of NAc MSNs subsequent to fear conditioning in mice. We then utilized an adeno-associated virus (AAV) transfection system to overexpress the SK3 subunit and explore the function of the NAcS MSNs SK3 channel in conditioned fear freezing. RESULTS Fear conditioning activated NAcS MSNs with enhanced excitability and reduced the SK channel-mediated medium after-hyperpolarization (mAHP) amplitude. The expression of NAcS SK3 were also reduced time-dependently. The overexpression of NAcS SK3 impaired conditioned fear consolidation without affecting conditioned fear expression, and blocked fear conditioning-induced alterations in NAcS MSNs excitability and mAHP amplitude. Additionally, the amplitudes of mEPSC, AMPAR/NMDAR ratio, and membrane surface GluA1/A2 expression in NAcS MSNs was increased by fear conditioning and returned to normal levels upon SK3 overexpression, indicating that fear conditioning-induced decrease of SK3 expression caused postsynaptic excitation by facilitating AMPAR transmission to the membrane. CONCLUSION These findings show that the NAcS MSNs SK3 channel plays a critical role in conditioned fear consolidation and that it may influence PTSD pathogenesis, making it a potential therapeutic target against PTSD.
Collapse
Affiliation(s)
- Minglong Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Province, Shijiazhuang, 050017, PR China
| | - Yixiao Luo
- Hunan Province People's Hospital, The First-Affiliated Hospital of Hunan Normal University, Changsha, 410081, PR China
| | - Jian Wang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Province, Shijiazhuang, 050017, PR China
| | - Yufei Sun
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Province, Shijiazhuang, 050017, PR China
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Province, Shijiazhuang, 050017, PR China
| | - Ludi Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Province, Shijiazhuang, 050017, PR China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Province, Shijiazhuang, 050017, PR China
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Province, Shijiazhuang, 050017, PR China.
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Province, Shijiazhuang, 050017, PR China.
| |
Collapse
|
25
|
Chapman DP, Vicini S, Burns MP, Evans R. Single Neuron Modeling Identifies Potassium Channel Modulation as Potential Target for Repetitive Head Impacts. Neuroinformatics 2023; 21:501-516. [PMID: 37294503 PMCID: PMC10833395 DOI: 10.1007/s12021-023-09633-7] [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] [Accepted: 05/17/2023] [Indexed: 06/10/2023]
Abstract
Traumatic brain injury (TBI) and repetitive head impacts can result in a wide range of neurological symptoms. Despite being the most common neurological disorder in the world, repeat head impacts and TBI do not have any FDA-approved treatments. Single neuron modeling allows researchers to extrapolate cellular changes in individual neurons based on experimental data. We recently characterized a model of high frequency head impact (HFHI) with a phenotype of cognitive deficits associated with decreases in neuronal excitability of CA1 neurons and synaptic changes. While the synaptic changes have been interrogated in vivo, the cause and potential therapeutic targets of hypoexcitability following repetitive head impacts are unknown. Here, we generated in silico models of CA1 pyramidal neurons from current clamp data of control mice and mice that sustained HFHI. We use a directed evolution algorithm with a crowding penalty to generate a large and unbiased population of plausible models for each group that approximated the experimental features. The HFHI neuron model population showed decreased voltage gated sodium conductance and a general increase in potassium channel conductance. We used partial least squares regression analysis to identify combinations of channels that may account for CA1 hypoexcitability after HFHI. The hypoexcitability phenotype in models was linked to A- and M-type potassium channels in combination, but not by any single channel correlations. We provide an open access set of CA1 pyramidal neuron models for both control and HFHI conditions that can be used to predict the effects of pharmacological interventions in TBI models.
Collapse
Affiliation(s)
- Daniel P Chapman
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Stefano Vicini
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Mark P Burns
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA.
- Department of Neuroscience, Georgetown University Medical Center, New Research Building-EG11, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA.
| | - Rebekah Evans
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA.
- Department of Neuroscience, Georgetown University Medical Center, New Research Building-EG11, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA.
| |
Collapse
|
26
|
Baracaldo-Santamaría D, Avendaño-Lopez SS, Ariza-Salamanca DF, Rodriguez-Giraldo M, Calderon-Ospina CA, González-Reyes RE, Nava-Mesa MO. Role of Calcium Modulation in the Pathophysiology and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24109067. [PMID: 37240413 DOI: 10.3390/ijms24109067] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease and the most frequent cause of progressive dementia in senior adults. It is characterized by memory loss and cognitive impairment secondary to cholinergic dysfunction and N-methyl-D-aspartate (NMDA)-mediated neurotoxicity. Intracellular neurofibrillary tangles, extracellular plaques composed of amyloid-β (Aβ), and selective neurodegeneration are the anatomopathological hallmarks of this disease. The dysregulation of calcium may be present in all the stages of AD, and it is associated with other pathophysiological mechanisms, such as mitochondrial failure, oxidative stress, and chronic neuroinflammation. Although the cytosolic calcium alterations in AD are not completely elucidated, some calcium-permeable channels, transporters, pumps, and receptors have been shown to be involved at the neuronal and glial levels. In particular, the relationship between glutamatergic NMDA receptor (NMDAR) activity and amyloidosis has been widely documented. Other pathophysiological mechanisms involved in calcium dyshomeostasis include the activation of L-type voltage-dependent calcium channels, transient receptor potential channels, and ryanodine receptors, among many others. This review aims to update the calcium-dysregulation mechanisms in AD and discuss targets and molecules with therapeutic potential based on their modulation.
Collapse
Affiliation(s)
- Daniela Baracaldo-Santamaría
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Sara Sofia Avendaño-Lopez
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Daniel Felipe Ariza-Salamanca
- Medical and Health Sciences Education Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Mateo Rodriguez-Giraldo
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| | - Carlos A Calderon-Ospina
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
- Grupo de Investigación en Ciencias Biomédicas Aplicadas (UR Biomed), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| | - Mauricio O Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| |
Collapse
|
27
|
Younes S, Mourad N, Salla M, Rahal M, Hammoudi Halat D. Potassium Ion Channels in Glioma: From Basic Knowledge into Therapeutic Applications. MEMBRANES 2023; 13:434. [PMID: 37103862 PMCID: PMC10144598 DOI: 10.3390/membranes13040434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 06/19/2023]
Abstract
Ion channels, specifically those controlling the flux of potassium across cell membranes, have recently been shown to exhibit an important role in the pathophysiology of glioma, the most common primary central nervous system tumor with a poor prognosis. Potassium channels are grouped into four subfamilies differing by their domain structure, gating mechanisms, and functions. Pertinent literature indicates the vital functions of potassium channels in many aspects of glioma carcinogenesis, including proliferation, migration, and apoptosis. The dysfunction of potassium channels can result in pro-proliferative signals that are highly related to calcium signaling as well. Moreover, this dysfunction can feed into migration and metastasis, most likely by increasing the osmotic pressure of cells allowing the cells to initiate the "escape" and "invasion" of capillaries. Reducing the expression or channel blockage has shown efficacy in reducing the proliferation and infiltration of glioma cells as well as inducing apoptosis, priming several approaches to target potassium channels in gliomas pharmacologically. This review summarizes the current knowledge on potassium channels, their contribution to oncogenic transformations in glioma, and the existing perspectives on utilizing them as potential targets for therapy.
Collapse
Affiliation(s)
- Samar Younes
- Department of Biomedical Sciences, School of Pharmacy, Lebanese International University, Bekaa 146404, Lebanon
- Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban (INSPECT-LB), Beirut 1103, Lebanon;
| | - Nisreen Mourad
- Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban (INSPECT-LB), Beirut 1103, Lebanon;
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese International University, Bekaa 146404, Lebanon; (M.R.)
| | - Mohamed Salla
- Department of Biological and Chemical Sciences, School of Arts and Sciences, Lebanese International University, Bekaa 146404, Lebanon;
| | - Mohamad Rahal
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese International University, Bekaa 146404, Lebanon; (M.R.)
| | - Dalal Hammoudi Halat
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese International University, Bekaa 146404, Lebanon; (M.R.)
- Academic Quality Department, QU Health, Qatar University, Doha 2713, Qatar;
| |
Collapse
|
28
|
Vera OD, Wulff H, Braun AP. Endothelial KCa channels: Novel targets to reduce atherosclerosis-driven vascular dysfunction. Front Pharmacol 2023; 14:1151244. [PMID: 37063294 PMCID: PMC10102451 DOI: 10.3389/fphar.2023.1151244] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023] Open
Abstract
Elevated levels of cholesterol in the blood can induce endothelial dysfunction, a condition characterized by impaired nitric oxide production and decreased vasodilatory capacity. Endothelial dysfunction can promote vascular disease, such as atherosclerosis, where macrophages accumulate in the vascular intima and fatty plaques form that impair normal blood flow in conduit arteries. Current pharmacological strategies to treat atherosclerosis mostly focus on lipid lowering to prevent high levels of plasma cholesterol that induce endothelial dysfunction and atherosclerosis. While this approach is effective for most patients with atherosclerosis, for some, lipid lowering is not enough to reduce their cardiovascular risk factors associated with atherosclerosis (e.g., hypertension, cardiac dysfunction, stroke, etc.). For such patients, additional strategies targeted at reducing endothelial dysfunction may be beneficial. One novel strategy to restore endothelial function and mitigate atherosclerosis risk is to enhance the activity of Ca2+-activated K+ (KCa) channels in the endothelium with positive gating modulator drugs. Here, we review the mechanism of action of these small molecules and discuss their ability to improve endothelial function. We then explore how this strategy could mitigate endothelial dysfunction in the context of atherosclerosis by examining how KCa modulators can improve cardiovascular function in other settings, such as aging and type 2 diabetes. Finally, we consider questions that will need to be addressed to determine whether KCa channel activation could be used as a long-term add-on to lipid lowering to augment atherosclerosis treatment, particularly in patients where lipid-lowering is not adequate to improve their cardiovascular health.
Collapse
Affiliation(s)
- O. Daniel Vera
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, United States
| | - Andrew P. Braun
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- *Correspondence: Andrew P. Braun,
| |
Collapse
|
29
|
Yoosefian M, Tajadini B, Ahmadzadeh S, Zeraati-Moghani M, Pakdin-Parizi Z. Ethanol effects on L-type voltage-gated calcium channel performance. J Mol Liq 2023. [DOI: 10.1016/j.molliq.2023.121634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
|
30
|
Numata T, Sato-Numata K, Yoshino M. Intermediate conductance Ca 2+-activated potassium channels are activated by functional coupling with stretch-activated nonselective cation channels in cricket myocytes. FRONTIERS IN INSECT SCIENCE 2023; 2:1100671. [PMID: 38468799 PMCID: PMC10926553 DOI: 10.3389/finsc.2022.1100671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/20/2022] [Indexed: 03/13/2024]
Abstract
Cooperative gating of localized ion channels ranges from fine-tuning excitation-contraction coupling in muscle cells to controlling pace-making activity in the heart. Membrane deformation resulting from muscle contraction activates stretch-activated (SA) cation channels. The subsequent Ca2+ influx activates spatially localized Ca2+-sensitive K+ channels to fine-tune spontaneous muscle contraction. To characterize endogenously expressed intermediate conductance Ca2+-activated potassium (IK) channels and assess the functional relevance of the extracellular Ca2+ source leading to IK channel activity, we performed patch-clamp techniques on cricket oviduct myocytes and recorded single-channel data. In this study, we first investigated the identification of IK channels that could be distinguished from endogenously expressed large-conductance Ca2+-activated potassium (BK) channels by adding extracellular Ba2+. The single-channel conductance of the IK channel was 62 pS, and its activity increased with increasing intracellular Ca2+ concentration but was not voltage-dependent. These results indicated that IK channels are endogenously expressed in cricket oviduct myocytes. Second, the Ca2+ influx pathway that activates the IK channel was investigated. The absence of extracellular Ca2+ or the presence of Gd3+ abolished the activity of IK channels. Finally, we investigated the proximity between SA and IK channels. The removal of extracellular Ca2+, administration of Ca2+ to the microscopic region in a pipette, and application of membrane stretching stimulation increased SA channel activity, followed by IK channel activity. Membrane stretch-induced SA and IK channel activity were positively correlated. However, the emergence of IK channel activity and its increase in response to membrane mechanical stretch was not observed without Ca2+ in the pipette. These results strongly suggest that IK channels are endogenously expressed in cricket oviduct myocytes and that IK channel activity is regulated by neighboring SA channel activity. In conclusion, functional coupling between SA and IK channels may underlie the molecular basis of spontaneous rhythmic contractions.
Collapse
Affiliation(s)
- Tomohiro Numata
- Department of Integrative Physiology, Graduate School of Medicine, Akita University, Akita, Japan
- Department of Biology, Tokyo Gakugei University, Tokyo, Japan
| | - Kaori Sato-Numata
- Department of Integrative Physiology, Graduate School of Medicine, Akita University, Akita, Japan
- Department of Biology, Tokyo Gakugei University, Tokyo, Japan
| | - Masami Yoshino
- Department of Biology, Tokyo Gakugei University, Tokyo, Japan
| |
Collapse
|
31
|
Ramis R, Ballesteros ÓR, Muguruza-Montero A, M-Alicante S, Núñez E, Villarroel Á, Leonardo A, Bergara A. Molecular dynamics simulations of the calmodulin-induced α-helix in the SK2 calcium-gated potassium ion channel. J Biol Chem 2022; 299:102850. [PMID: 36587765 PMCID: PMC9874072 DOI: 10.1016/j.jbc.2022.102850] [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: 06/17/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
The family of small-conductance Ca2+-activated potassium ion channels (SK channels) is composed of four members (SK1, SK2, SK3, and SK4) involved in neuron-firing regulation. The gating of these channels depends on the intracellular Ca2+ concentration, and their sensitivity to this ion is provided by calmodulin (CaM). This protein binds to a specific region in SK channels known as the calmodulin-binding domain (CaMBD), an event which is essential for their gating. While CaMBDs are typically disordered in the absence of CaM, the SK2 channel subtype displays a small prefolded α-helical region in its CaMBD even if CaM is not present. This small helix is known to turn into a full α-helix upon CaM binding, although the molecular-level details for this conversion are not fully understood yet. In this work, we offer new insights on this physiologically relevant process by means of enhanced sampling, atomistic Hamiltonian replica exchange molecular dynamics simulations, providing a more detailed understanding of CaM binding to this target. Our results show that CaM is necessary for inducing a full α-helix along the SK2 CaMBD through hydrophobic interactions with V426 and L427. However, it is also necessary that W431 does not compete for these interactions; the role of the small prefolded α-helix in the SK2 CaMBD would be to stabilize W431 so that this is the case. In conclusion, our findings provide further insight into a key interaction between CaM and SK channels that is important for channel sensitivity to Ca2+.
Collapse
Affiliation(s)
- Rafael Ramis
- Donostia International Physics Center, Donostia, Spain; Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain.
| | - Óscar R. Ballesteros
- Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain,Centro de Física de Materiales CFM, CSIC-UPV/EHU, Donostia, Spain
| | | | - Sara M-Alicante
- Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain,Instituto Biofisika, CSIC-UPV/EHU, Leioa, Spain
| | - Eider Núñez
- Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain,Instituto Biofisika, CSIC-UPV/EHU, Leioa, Spain
| | | | - Aritz Leonardo
- Donostia International Physics Center, Donostia, Spain,Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain
| | - Aitor Bergara
- Donostia International Physics Center, Donostia, Spain,Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain,Centro de Física de Materiales CFM, CSIC-UPV/EHU, Donostia, Spain
| |
Collapse
|
32
|
Butler AS, Hancox JC, Marrion NV. Preferential formation of human heteromeric SK2:SK3 channels limits homomeric SK channel assembly and function. J Biol Chem 2022; 299:102783. [PMID: 36502918 PMCID: PMC9841042 DOI: 10.1016/j.jbc.2022.102783] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/29/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
Three isoforms of small conductance, calcium-activated potassium (SK) channel subunits have been identified (SK1-3) that exhibit a broad and overlapping tissue distribution. SK channels have been implicated in several disease states including hypertension and atrial fibrillation, but therapeutic targeting of SK channels is hampered by a lack of subtype-selective inhibitors. This is further complicated by studies showing that SK1 and SK2 preferentially form heteromeric channels during co-expression, likely limiting the function of homomeric channels in vivo. Here, we utilized a simplified expression system to investigate functional current produced when human (h) SK2 and hSK3 subunits are co-expressed. When expressed alone, hSK3 subunits were more clearly expressed on the cell surface than hSK2 subunits. hSK3 surface expression was reduced by co-transfection with hSK2. Whole-cell recording showed homomeric hSK3 currents were larger than homomeric hSK2 currents or heteromeric hSK2:hSK3 currents. The smaller amplitude of hSK2:hSK3-mediated current when compared with homomeric hSK3-mediated current suggests hSK2 subunits regulate surface expression of heteromers. Co-expression of hSK2 and hSK3 subunits produced a current that arose from a single population of heteromeric channels as exhibited by an intermediate sensitivity to the inhibitors apamin and UCL1684. Co-expression of the apamin-sensitive hSK2 subunit and a mutant, apamin-insensitive hSK3 subunit [hSK3(H485N)], produced an apamin-sensitive current. Concentration-inhibition relationships were best fit by a monophasic Hill equation, confirming preferential formation of heteromers. These data show that co-expressed hSK2 and hSK3 preferentially form heteromeric channels and suggest that the hSK2 subunit acts as a chaperone, limiting membrane expression of hSK2:hSK3 heteromeric channels.
Collapse
Affiliation(s)
- Andrew S Butler
- School of Physiology, Pharmacology and Neuroscience, Medical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom
| | - Jules C Hancox
- School of Physiology, Pharmacology and Neuroscience, Medical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom.
| | - Neil V Marrion
- School of Physiology, Pharmacology and Neuroscience, Medical Sciences Building, University of Bristol, University Walk, Bristol, United Kingdom.
| |
Collapse
|
33
|
Lyu S, Xing H, Liu Y, Girdhar P, Yokoi F, Li Y. Further Studies on the Role of BTBD9 in the Cerebellum, Sleep-like Behaviors and the Restless Legs Syndrome. Neuroscience 2022; 505:78-90. [PMID: 36244636 PMCID: PMC10367443 DOI: 10.1016/j.neuroscience.2022.10.008] [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: 08/05/2022] [Revised: 09/15/2022] [Accepted: 10/09/2022] [Indexed: 11/26/2022]
Abstract
Genetic analyses have linked BTBD9 to restless legs syndrome (RLS) and sleep regulation. Btbd9 knockout mice show RLS-like motor restlessness. Previously, we found hyperactivity of cerebellar Purkinje cells (PCs) in Btbd9 knockout mice, which may contribute to the motor restlessness observed. However, underlying mechanisms for PC hyperactivity in Btbd9 knockout mice are unknown. Here, we used dissociated PC recording, brain slice recording and western blot to address this question. Our dissociated recording shows that knockout PCs had increased TEA-sensitive, Ca2+-dependent K+ currents. Applying antagonist to large conductance Ca2+-activated K+ (BK) channels further isolated the increased current as BK current. Consistently, we found increased amplitude of afterhyperpolarization and elevated BK protein levels in the knockout mice. Dissociated recording also shows a decrease in TEA-insensitive, Ca2+-dependent K+ currents. The result is consistent with reduced amplitude of tail currents, mainly composed of small conductance Ca2+-activated K+ (SK) currents, in slice recording. Our results suggest that BK and SK channels may be responsible for the hyperactivity of knockout PCs. Recently, BTBD9 protein was shown to associate with SYNGAP1 protein. We found a decreased cerebellar level of SYNGAP1 in Btbd9 knockout mice. However, Syngap1 heterozygous knockout mice showed nocturnal, instead of diurnal, motor restlessness. Our results suggest that SYNGAP1 deficiency may not contribute directly to the RLS-like motor restlessness observed in Btbd9 knockout mice. Finally, we found that PC-specific Btbd9 knockout mice exhibited deficits in motor coordination and balance similar to Btbd9 knockout mice, suggesting that the motor effect of BTBD9 in PCs is cell-autonomous.
Collapse
Affiliation(s)
- Shangru Lyu
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Hong Xing
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yuning Liu
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Pallavi Girdhar
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Fumiaki Yokoi
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yuqing Li
- Norman Fixel Institute for Neurological Diseases, Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
34
|
External Hemin as an Inhibitor of Mitochondrial Large-Conductance Calcium-Activated Potassium Channel Activity. Int J Mol Sci 2022; 23:ijms232113391. [DOI: 10.3390/ijms232113391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/26/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
The mitochondrial large-conductance calcium-activated potassium channel (mitoBKCa) is located in the inner mitochondrial membrane and seems to play a crucial role in cytoprotection. The mitoBKCa channel is regulated by many modulators, including activators, such as calcium ions and inhibitors, such as heme and its oxidized form hemin. Heme/hemin binds to the heme-binding motif (CXXCH) located between two RCK domains present in the mitochondrial matrix. In the present study, we used the patch-clamp technique in the outside-out configuration to record the activity of mitoBKCa channels. This allowed for the application of channel modulators to the intermembrane-space side of the mitoBKCa. We found that hemin applied in this configuration inhibits the activity of mitoBKCa. In addition, we proved that the observed hemin effect is specific and it is not due to its interaction with the inner mitochondrial membrane. Our data suggest the existence of a new potential heme/hemin binding site in the structure of the mitoBKCa channel located on the mitochondrial intermembrane space side, which could constitute a new way for the regulation of mitoBKCa channel activity.
Collapse
|
35
|
Leitner DF, Devore S, Laze J, Friedman D, Mills JD, Liu Y, Janitz M, Anink JJ, Baayen JC, Idema S, van Vliet EA, Diehl B, Scott C, Thijs R, Nei M, Askenazi M, Sivathamboo S, O’Brien T, Wisniewski T, Thom M, Aronica E, Boldrini M, Devinsky O. Serotonin receptor expression in hippocampus and temporal cortex of temporal lobe epilepsy patients by postictal generalized electroencephalographic suppression duration. Epilepsia 2022; 63:2925-2936. [PMID: 36053862 PMCID: PMC9669210 DOI: 10.1111/epi.17400] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/18/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Prolonged postictal generalized electroencephalographic suppression (PGES) is a potential biomarker for sudden unexpected death in epilepsy (SUDEP), which may be associated with dysfunctional autonomic responses and serotonin signaling. To better understand molecular mechanisms, PGES duration was correlated to 5HT1A and 5HT2A receptor protein expression and RNAseq from resected hippocampus and temporal cortex of temporal lobe epilepsy patients with seizures recorded in preoperative evaluation. METHODS Analyses included 36 cases (age = 14-64 years, age at epilepsy onset = 0-51 years, epilepsy duration = 2-53 years, PGES duration = 0-93 s), with 13 cases in all hippocampal analyses. 5HT1A and 5HT2A protein was evaluated by Western blot and histologically in hippocampus (n = 16) and temporal cortex (n = 9). We correlated PGES duration to our previous RNAseq dataset for serotonin receptor expression and signaling pathways, as well as weighted gene correlation network analysis (WGCNA) to identify correlated gene clusters. RESULTS In hippocampus, 5HT2A protein by Western blot positively correlated with PGES duration (p = .0024, R2 = .52), but 5HT1A did not (p = .87, R2 = .0020). In temporal cortex, 5HT1A and 5HT2A had lower expression and did not correlate with PGES duration. Histologically, PGES duration did not correlate with 5HT1A or 5HT2A expression in hippocampal CA4, dentate gyrus, or temporal cortex. RNAseq identified two serotonin receptors with expression that correlated with PGES duration in an exploratory analysis: HTR3B negatively correlated (p = .043, R2 = .26) and HTR4 positively correlated (p = .049, R2 = .25). WGCNA identified four modules correlated with PGES duration, including positive correlation with synaptic transcripts (p = .040, Pearson correlation r = .52), particularly potassium channels (KCNA4, KCNC4, KCNH1, KCNIP4, KCNJ3, KCNJ6, KCNK1). No modules were associated with serotonin receptor signaling. SIGNIFICANCE Higher hippocampal 5HT2A receptor protein and potassium channel transcripts may reflect underlying mechanisms contributing to or resulting from prolonged PGES. Future studies with larger cohorts should assess functional analyses and additional brain regions to elucidate mechanisms underlying PGES and SUDEP risk.
Collapse
Affiliation(s)
- Dominique F. Leitner
- Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY, USA
- Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Sasha Devore
- Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Juliana Laze
- Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Daniel Friedman
- Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - James D. Mills
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
- Department of Clinical and Experimental Epilepsy, University College London Institute of Neurology, London, UK
- Chalfont Centre for Epilepsy, Bucks, UK
| | - Yan Liu
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, USA
| | - Michael Janitz
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Jasper J. Anink
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
| | - Johannes C. Baayen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Sander Idema
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Neurosurgery, Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Erwin A. van Vliet
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Beate Diehl
- Department of Clinical and Experimental Epilepsy, University College London Institute of Neurology, London, UK
| | - Catherine Scott
- Department of Clinical and Experimental Epilepsy, University College London Institute of Neurology, London, UK
| | - Roland Thijs
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands
| | - Maromi Nei
- Department of Neurology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Manor Askenazi
- Biomedical Hosting LLC, Arlington, MA, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Shobi Sivathamboo
- Department of Neuroscience, Alfred Health, Central Clinical School, Melbourne, Victoria, Australia
- Department Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Terence O’Brien
- Department of Neuroscience, Alfred Health, Central Clinical School, Melbourne, Victoria, Australia
- Department Medicine, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
| | - Thomas Wisniewski
- Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - Maria Thom
- Department of Clinical and Experimental Epilepsy, University College London Institute of Neurology, London, UK
| | - Eleonora Aronica
- Amsterdam UMC location University of Amsterdam, Department of (Neuro)pathology, Amsterdam Neuroscience, Meibergdreef 9, Amsterdam, the Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands
| | - Maura Boldrini
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Orrin Devinsky
- Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY, USA
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
36
|
Cahusac PM, Senok SS. Effects of potassium channel modulators on the responses of mammalian slowly adapting mechanoreceptors. IBRO Neurosci Rep 2022; 13:344-355. [PMID: 36274789 PMCID: PMC9582710 DOI: 10.1016/j.ibneur.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 10/06/2022] [Indexed: 11/08/2022] Open
Abstract
Introduction slowly adapting mechanoreceptors in the skin provide vital tactile information to animals. The ionic channels that underlie their functioning is the subject of intense research. Previous work suggests that potassium channels may play particular roles in the activation and firing of these mechanoreceptors. Objective We used a range of potassium channel blockers and openers to observe their effects on different phases of mechanoreceptor responses. Methods Extracellular recording of neural activity of slowly adapting mechanoreceptors was carried out in an in vitro preparation of the sinus hair follicles taken from rat whisker pads. A range of potassium (K+) channel modulators were tested on these mechanoreceptor responses. The channel blockers tested were: tetraethylammonium (TEA), barium chloride (BaCl2), dequalinium, 4-aminopyridine (4-AP), paxilline, XE 991, apamin, and charybdotoxin. Results Except for charybdotoxin and apamin, these drugs increased the activity of both types of slowly adapting units, St I and St II. Generally, both spontaneous and evoked (dynamic and static) activities increased. The channel opener NS1619 was also tested. NS1619 clearly decreased evoked activity (both dynamic and static) while leaving spontaneous activity relatively unaffected, with no clear discrimination of effects on the two types of St receptor Conclusion These findings are consistent with the targets of the drugs suggesting that K+ channels play an important role in the maintenance of spontaneous firing and in the production of and persistence of mechanoreceptor activity.
Collapse
Affiliation(s)
- Peter M.B. Cahusac
- College of Medicine, Alfaisal University, Saudi Arabia, and Department of Comparative Medicine, King Faisal Specialist Hospital & Research Centre, PO Box 50927, Riyadh 11533, Kingdom of Saudi Arabia,Correspondence to: Department of Pharmacology & Biostatistics College of Medicine Alfaisal University, PO Box 50927, Riyadh 11533, Kingdom of Saudi Arabia.
| | - Solomon S. Senok
- Ajman University College of Medicine, PO Box 346, Ajman, United Arab Emirates
| |
Collapse
|
37
|
Boyle Y, Johns TG, Fletcher EV. Potassium Ion Channels in Malignant Central Nervous System Cancers. Cancers (Basel) 2022; 14:cancers14194767. [PMID: 36230692 PMCID: PMC9563970 DOI: 10.3390/cancers14194767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Malignant central nervous system (CNS) cancers are among the most difficult to treat, with low rates of survival and a high likelihood of recurrence. This is primarily due to their location within the CNS, hindering adequate drug delivery and tumour access via surgery. Furthermore, CNS cancer cells are highly plastic, an adaptive property that enables them to bypass targeted treatment strategies and develop drug resistance. Potassium ion channels have long been implicated in the progression of many cancers due to their integral role in several hallmarks of the disease. Here, we will explore this relationship further, with a focus on malignant CNS cancers, including high-grade glioma (HGG). HGG is the most lethal form of primary brain tumour in adults, with the majority of patient mortality attributed to drug-resistant secondary tumours. Hence, targeting proteins that are integral to cellular plasticity could reduce tumour recurrence, improving survival. This review summarises the role of potassium ion channels in malignant CNS cancers, specifically how they contribute to proliferation, invasion, metastasis, angiogenesis, and plasticity. We will also explore how specific modulation of these proteins may provide a novel way to overcome drug resistance and improve patient outcomes.
Collapse
Affiliation(s)
- Yasmin Boyle
- Telethon Kids Institute, Perth Children’s Hospital, 15 Hospital Ave, Nedlands, Perth, WA 6009, Australia
- School of Biomedicine, The University of Western Australia, 35 Stirling Hwy, Crawley, Perth, WA 6009, Australia
- Correspondence:
| | - Terrance G. Johns
- Telethon Kids Institute, Perth Children’s Hospital, 15 Hospital Ave, Nedlands, Perth, WA 6009, Australia
- School of Biomedicine, The University of Western Australia, 35 Stirling Hwy, Crawley, Perth, WA 6009, Australia
| | - Emily V. Fletcher
- Telethon Kids Institute, Perth Children’s Hospital, 15 Hospital Ave, Nedlands, Perth, WA 6009, Australia
- School of Biomedicine, The University of Western Australia, 35 Stirling Hwy, Crawley, Perth, WA 6009, Australia
| |
Collapse
|
38
|
King DR, Sedovy MW, Eaton X, Dunaway LS, Good ME, Isakson BE, Johnstone SR. Cell-To-Cell Communication in the Resistance Vasculature. Compr Physiol 2022; 12:3833-3867. [PMID: 35959755 DOI: 10.1002/cphy.c210040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The arterial vasculature can be divided into large conduit arteries, intermediate contractile arteries, resistance arteries, arterioles, and capillaries. Resistance arteries and arterioles primarily function to control systemic blood pressure. The resistance arteries are composed of a layer of endothelial cells oriented parallel to the direction of blood flow, which are separated by a matrix layer termed the internal elastic lamina from several layers of smooth muscle cells oriented perpendicular to the direction of blood flow. Cells within the vessel walls communicate in a homocellular and heterocellular fashion to govern luminal diameter, arterial resistance, and blood pressure. At rest, potassium currents govern the basal state of endothelial and smooth muscle cells. Multiple stimuli can elicit rises in intracellular calcium levels in either endothelial cells or smooth muscle cells, sourced from intracellular stores such as the endoplasmic reticulum or the extracellular space. In general, activation of endothelial cells results in the production of a vasodilatory signal, usually in the form of nitric oxide or endothelial-derived hyperpolarization. Conversely, activation of smooth muscle cells results in a vasoconstriction response through smooth muscle cell contraction. © 2022 American Physiological Society. Compr Physiol 12: 1-35, 2022.
Collapse
Affiliation(s)
- D Ryan King
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA
| | - Meghan W Sedovy
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA.,Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Blacksburg, Virginia, USA
| | - Xinyan Eaton
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA
| | - Luke S Dunaway
- Robert M. Berne Cardiovascular Research Centre, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Miranda E Good
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Centre, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Scott R Johnstone
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
39
|
Alaee E, Farahani F, Semnanian S, Azizi H. Prenatal exposure to morphine enhances excitability in locus coeruleus neurons. J Neural Transm (Vienna) 2022; 129:1049-1060. [PMID: 35674919 DOI: 10.1007/s00702-022-02515-3] [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: 03/28/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
Abstract
Opioid abuse during pregnancy may have noteworthy effects on the child's behavioral, emotional and cognitive progression. In this study, we assessed the effect of prenatal exposure to morphine on electrophysiological features of locus coeruleus (LC) noradrenergic neurons which is involved in modulating cognitive performance. Pregnant dams were randomly divided into two groups, that is a prenatal saline treated and prenatal morphine-treated group. To this end, on gestational days 11-18, either morphine or saline (twice daily, s.c.) was administered to pregnant dams. Whole-cell patch-clamp recordings were conducted on LC neurons of male offspring. The evoked firing rate, instantaneous frequency and action potentials half-width, and also input resistance of LC neurons significantly increased in the prenatal morphine group compared to the saline group. Moreover, action potentials decay slope, after hyperpolarization amplitude, rheobase current, and first spike latency were diminished in LC neurons following prenatal exposure to morphine. In addition, resting membrane potential, rise slope, and amplitude of action potentials were not changed by prenatal morphine exposure. Together, the current findings show a significant enhancement in excitability of the LC neurons following prenatal morphine exposure, which may affect the release of norepinephrine to other brain regions and/or cognitive performances of the offspring.
Collapse
Affiliation(s)
- Elham Alaee
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Farahani
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeed Semnanian
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Azizi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
40
|
Alza L, Visa A, Herreros J, Cantí C. T-type channels in cancer cells: Driving in reverse. Cell Calcium 2022; 105:102610. [PMID: 35691056 DOI: 10.1016/j.ceca.2022.102610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/01/2022] [Accepted: 06/04/2022] [Indexed: 11/30/2022]
Abstract
In the strongly polarized membranes of excitable cells, activation of T-type Ca2+ channels (TTCCs) by weak depolarizing stimuli allows the influx of Ca2+ which further amplifies membrane depolarization, thus "recruiting" higher threshold voltage-gated channels to promote action potential firing. Nonetheless, TTCCs perform other functions in the plasma membrane of both excitable and non-excitable cells, in which they regulate a number of biochemical pathways relevant for cell cycle and cell fate. Furthermore, data obtained in the last 20 years have shown the involvement of TTCCs in tumor biology, designating them as promising chemotherapeutic targets. However, their activity in the steadily-depolarized membranes of cancer cells, in which most voltage-gated channels are in the inactivated (nonconducting) state, is counter-intuitive. Here we discuss that in cancer cells weak hyperpolarizing stimuli increase the fraction of open TTCCs which, in association with Ca2+-dependent K+ channels, may critically boost membrane hyperpolarization and driving force for Ca2+ entry through different voltage-independent Ca2+ channels. Available evidence also shows that TTCCs participate in positive feedback circuits with signaling effectors, which may warrant a switch-like activation of pro-proliferative and pro-survival pathways in spite of their low availability. Unravelling TTCC modus operandi in the context of non-excitable membranes may facilitate the development of novel anticancer approaches.
Collapse
Affiliation(s)
- Lía Alza
- Universitat de Lleida (Dpt. Medicina Experimental), IRBLleida, Rovira Roure 80, Lleida 25198, Spain
| | - Anna Visa
- Universitat de Lleida (Dpt. Medicina Experimental), IRBLleida, Rovira Roure 80, Lleida 25198, Spain
| | - Judit Herreros
- Universitat de Lleida (Dpt. Ciències Mèdiques Bàsiques), IRBLleida
| | - Carles Cantí
- Universitat de Lleida (Dpt. Medicina Experimental), IRBLleida, Rovira Roure 80, Lleida 25198, Spain.
| |
Collapse
|
41
|
Zuccolini P, Gavazzo P, Pusch M. BK Channel in the Physiology and in the Cancer of Pancreatic Duct: Impact and Reliability of BK Openers. Front Pharmacol 2022; 13:906608. [PMID: 35685628 PMCID: PMC9171006 DOI: 10.3389/fphar.2022.906608] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
BK (KCa 1.1, Slo-1) is a K+ channel characterized by an allosteric regulation of the gating mechanism by Ca2+ binding and voltage, and a high unitary conductance. The channel is expressed in many different tissues, where it is involved in the regulation or the fine-tuning of many physiological processes. Among other organs, BK is expressed in the pancreatic duct, a part of the gland important for the correct ionic composition of the pancreatic juice. Unfortunately, the pancreatic duct is also the site where one of the deadliest cancer types, the pancreatic duct adenocarcinoma (PDAC), develops. In the past years, it has been reported that continuous exposure of cancer cells to BK openers can have a significant impact on cell viability as well as on the ability to proliferate and migrate. Here, we first summarize the main BK channel properties and its roles in pancreatic duct physiology. Then we focus on the potential role of BK as a pharmacological target in PDAC. Moreover, we discuss how results obtained when employing BK activators on cancer cells can, in some cases, be misleading.
Collapse
|
42
|
A novel role of BK potassium channel activity in preventing the development of kidney fibrosis. Kidney Int 2022; 101:945-962. [PMID: 34968553 DOI: 10.1016/j.kint.2021.11.033] [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] [Received: 07/26/2021] [Revised: 11/03/2021] [Accepted: 11/22/2021] [Indexed: 11/21/2022]
Abstract
Kidney fibrosis is a common characteristic of chronic kidney disease and while the large conductance voltage and calcium-activated potassium channel (BK) is widely expressed in kidneys, its role in kidney fibrosis is unknown. To evaluate this, we found that BK protein expression was decreased in the fibrotic kidneys. Accompanying this was increased fibrotic marker protein expression of fibronectin, vimentin and α-smooth muscle actin and increased mRNA expressions of fibronectin, α-smooth muscle actin, collagen III and collagen I. These changes occurred in the unilateral ureteral obstruction and folic acid models of fibrosis and were more pronounced in BK knockout than in wild-type mice. Activation of BK activity by chemical NS1619 or BMS191011 channel openers attenuated kidney fibrosis in these two models while protecting kidney function in wild-type mice. BK deficiency up-regulated transforming growth factor-β (TGF-β)/transcription factor Smad2/3 signaling in the fibrotic kidney, whereas activation of BK activity inhibited this signaling pathway both in vivo and in vitro. BK channel activation increased the degradation of TGF-β receptors induced by TGF-β1 in vivo and in vitro. Furthermore, in cell lines HK-2, NRK49, and NRK-52E, BK channel activation by NS1619 led to increased caveolae formation and facilitated localization of TGF-β receptors in the microdomains of lipid rafts. Thus, our data demonstrated that BK activation has an anti-fibrotic effect on kidney fibrosis by inhibiting the TGF-β signaling pathway through accelerating TGF-β receptor degradation via the caveolae route. Hence, our study provides innovative insight into BK as a potential therapeutic target for the treatment of kidney fibrosis.
Collapse
|
43
|
Zhang D, Zhu Q, Xia W, Zhu C, Zhao X, Zhang Y, He C, Ji S, Li X, Zhang J. The role of SK3 in progesterone-induced inhibition of human fallopian tubal contraction. Reprod Biol Endocrinol 2022; 20:73. [PMID: 35488306 PMCID: PMC9052544 DOI: 10.1186/s12958-022-00932-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/22/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Normal motor activity of the fallopian tube is critical for human reproduction, and abnormal tubal activity may lead to ectopic pregnancy (EP) or infertility. Progesterone has an inhibitory effect on tubal contraction; however, the underlying mechanisms remain unclear. Small-conductance calcium-activated K+ channel 3 (SK3) is abundantly expressed in platelet-derived growth factor receptor α positive (PDGFRα+) cells and was reported to be important for the relaxation of smooth muscle. The present study aims to explore the expression of SK3 in the human fallopian tube and its role in progesterone-induced inhibition of tubal contraction. METHODS We collected specimens of fallopian tubes from patients treated by salpingectomy for EP (EP group) and other benign gynecological diseases (Non-EP group). The expression of SK3 was detected by quantitative real-time polymerase chain reaction, western blot, immunocytochemistry, and immunohistochemistry analyses. Isometric tension experiments were performed to investigate the role of SK3 in progesterone-induced inhibition of tubal contraction. RESULTS The baseline amplitude and frequency of human fallopian tube contraction were both statistically lower in the EP group compared with the non-EP group. The expression levels of SK3 in different portions of fallopian tubes from the non-EP group were significantly higher than in those from the EP group. Progesterone had an inhibitory effect on tubal contraction, mainly on the amplitude, in both groups, and SK3 as well as other calcium-activated K+ channels may be involved. SK3-expressing PDGFRα (+) cells were detected in the human fallopian tube. CONCLUSIONS The expression of SK3 is lower in the EP group, and SK3 is involved in the progesterone-induced inhibition of human fallopian tube contraction.
Collapse
Affiliation(s)
- Duo Zhang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Qian Zhu
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Wei Xia
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Chenfeng Zhu
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Xiaoya Zhao
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Yiqin Zhang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Chuqing He
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Sifan Ji
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200030, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China
| | - Xiaocui Li
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Jian Zhang
- Department of Obstetrics and Gynecology, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200030, China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, China.
| |
Collapse
|
44
|
Yin H, Cheng H, Li P, Yang Z. TRPC6 interacted with K Ca1.1 channels to regulate the proliferation and apoptosis of glioma cells. Arch Biochem Biophys 2022; 725:109268. [PMID: 35489424 DOI: 10.1016/j.abb.2022.109268] [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: 11/14/2021] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 11/17/2022]
Abstract
Malignant glioma is the most aggressive and deadliest brain malignancy. TRPC6 and KCa1.1, two ion channels, have been considered as potential therapeutic targets for malignant glioma treatment. TRPC6, a Ca2+-permeable channel, plays a vital role in promoting tumorigenesis and the progression of glioma. KCa1.1, a large-conductance Ca2+-activated channel, is also involved in growth and migration of glioma. However, the underlying mechanism by which these two ion channels promote glioma progression was unclear. In our study, we found that TRPC6 upregulated the expression of KCa1.1, while the immunoprecipitation analysis also showed that TRPC6 interacts with KCa1.1 channels in glioma cells. The currents of KCa1.1 recorded by the whole-cell patch clamp technique were increased by TRPC6 in glioma cells, suggesting that TRPC6 can provide a Ca2+ source for the activation of KCa1.1 channels. It was also suggested that TRPC6 regulates the proliferation and apoptosis of glioma cells through KCa1.1 channels in vitro. Therefore, C6-bearing glioma rats were established to validate the results in vitro. After the administration of paxilline (a specific inhibitor of KCa1.1 channels), TRPC6-dependent growth of glioma was inhibited in vivo. We also found that TRPC6 enhanced co-expression with KCa1.1 in glioma. These all suggested that TRPC6/KCa1.1 signal plays a role in promoting the growth of glioma. Our results provided new evidence for TRPC6 and KCa1.1 as potential targets for glioma treatment.
Collapse
Affiliation(s)
- Hongqiang Yin
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin, 300071, China; CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Haofeng Cheng
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Peiqi Li
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Zhuo Yang
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
45
|
Rajabian A, Rajabian F, Babaei F, Mirzababaei M, Nassiri-Asl M, Hosseinzadeh H. Interaction of Medicinal Plants and Their Active Constituents With Potassium Ion Channels: A Systematic Review. Front Pharmacol 2022; 13:831963. [PMID: 35273505 PMCID: PMC8902679 DOI: 10.3389/fphar.2022.831963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Potassium ion (K+) channels are pore-forming transmembrane proteins that control the transport of K+ ions. Medicinal plants are widely used as complementary therapies for several disorders. Studies have shown that the modulation of K+ channels is most likely involved in various pharmacological effects of medicinal plants. This review aimed to evaluate the modulatory effects of medicinal plants and their active constituents on K+ channels under pathological conditions. This systematic review was prepared according to the Preferred Reporting Items for the Systematic Reviews and Meta-analyses (PRISMA) 2020 guideline. Four databases, including PubMed, Web of Science, embase, and Scopus, were searched. We identified 687 studies from these databases, from which we selected 13 in vivo studies for the review by using the Population, Intervention, Comparison, Outcomes, Study (PICOS) tool. The results of the 13 selected studies showed a modulatory effect of medicinal plants or their active constituents on ATP-sensitive potassium channels (KATP), and small (SKCa) and large (BKCa) conductance calcium-activated K+ channels in several pathological conditions such as nociception, brain ischemia, seizure, diabetes, gastric ulcer, myocardial ischemia-reperfusion, and hypertension via possible involvement of the nitric oxide/cyclic GMP pathway and protein kinase. K+ channels should be considered as significant therapeutic milestones in the treatment of several diseases. We believe that understanding the mechanism behind the interaction of medicinal plants with K+ channels can facilitate drug development for the treatment of various K+ channel-related disorders.
Collapse
Affiliation(s)
- Arezoo Rajabian
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Rajabian
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Babaei
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Mirzababaei
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Marjan Nassiri-Asl
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
46
|
An unbiased algorithm for objective separation of Alzheimer's, Alzheimer's mixed with cerebrovascular symptomology, and healthy controls from one another using electrovestibulography (EVestG). Med Biol Eng Comput 2022; 60:797-810. [PMID: 35102489 DOI: 10.1007/s11517-022-02507-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/18/2022] [Indexed: 10/19/2022]
Abstract
Diagnosis of Alzheimer's disease (AD) from AD with cerebrovascular disease pathology (AD-CVD) is a rising challenge. Using electrovestibulography (EVestG) measured signals, we develop an automated feature extraction and selection algorithm for an unbiased identification of AD and AD-CVD from healthy controls as well as their separation from each other. EVestG signals of 24 healthy controls, 16 individuals with AD, and 13 with AD-CVD were analyzed within two separate groupings: One-versus-One and One-versus-All. A multistage feature selection process was conducted over the training dataset using linear support vector machine (SVM) classification with 10-fold cross-validation, k nearest neighbors/averaging imputation, and exhaustive search. The most frequently selected features that achieved highest classification performance were selected. 10-fold cross-validation was applied via a linear SVM classification on the entire dataset. Multivariate analysis was run to test the between population differences while controlling for the covariates. Classification accuracies of ≥ 80% and 78% were achieved for the One-versus-All classification approach and AD versus AD-CVD separation, respectively. The results also held true after controlling for the effect of covariates. AD/AD-CVD participants showed smaller/larger EVestG averaged field potential signals compared to healthy controls and AD-CVD/AD participants. These characteristics are in line with our previous study results.
Collapse
|
47
|
Onose G, Anghelescu A, Blendea D, Ciobanu V, Daia C, Firan FC, Oprea M, Spinu A, Popescu C, Ionescu A, Busnatu Ș, Munteanu C. Cellular and Molecular Targets for Non-Invasive, Non-Pharmacological Therapeutic/Rehabilitative Interventions in Acute Ischemic Stroke. Int J Mol Sci 2022; 23:907. [PMID: 35055089 PMCID: PMC8846361 DOI: 10.3390/ijms23020907] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cerebral circulation delivers the blood flow to the brain through a dedicated network of sanguine vessels. A healthy human brain can regulate cerebral blood flow (CBF) according to any physiological or pathological challenges. The brain is protected by its self-regulatory mechanisms, which are dependent on neuronal and support cellular populations, including endothelial ones, as well as metabolic, and even myogenic factors. OBJECTIVES Accumulating data suggest that "non-pharmacological" approaches might provide new opportunities for stroke therapy, such as electro-/acupuncture, hyperbaric oxygen therapy, hypothermia/cooling, photobiomodulation, therapeutic gases, transcranial direct current stimulations, or transcranial magnetic stimulations. We reviewed the recent data on the mechanisms and clinical implications of these non-pharmaceutical treatments. METHODS To present the state-of-the-art for currently available non-invasive, non-pharmacological-related interventions in acute ischemic stroke, we accomplished this synthetic and systematic literature review based on the Preferred Reporting Items for Systematic Principles Reviews and Meta-Analyses (PRISMA). RESULTS The initial number of obtained articles was 313. After fulfilling the five steps in the filtering/selection methodology, 54 fully eligible papers were selected for synthetic review. We enhanced our documentation with other bibliographic resources connected to our subject, identified in the literature within a non-standardized search, to fill the knowledge gaps. Fifteen clinical trials were also identified. DISCUSSION Non-invasive, non-pharmacological therapeutic/rehabilitative interventions for acute ischemic stroke are mainly holistic therapies. Therefore, most of them are not yet routinely used in clinical practice, despite some possible beneficial effects, which have yet to be supplementarily proven in more related studies. Moreover, few of the identified clinical trials are already completed and most do not have final results. CONCLUSIONS This review synthesizes the current findings on acute ischemic stroke therapeutic/rehabilitative interventions, described as non-invasive and non-pharmacological.
Collapse
Affiliation(s)
- Gelu Onose
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.D.); (M.O.); (A.S.); (A.I.); (Ș.B.)
- Neuromuscular Rehabilitation Clinic Division, Teaching Emergency Hospital” Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.A.); (C.P.)
| | - Aurelian Anghelescu
- Neuromuscular Rehabilitation Clinic Division, Teaching Emergency Hospital” Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.A.); (C.P.)
- Faculty of Midwives and Nursing, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Dan Blendea
- Faculty of Medicine, University ”Titu Maiorescu”, 0400511 Bucharest, Romania;
- Physical and Rehabilitation Medicine & Balneology Clinic Division, Teaching Emergency Hospital of the Ilfov County, 022113 Bucharest, Romania;
| | - Vlad Ciobanu
- Computer Science Department, Politehnica University of Bucharest, 060042 Bucharest, Romania;
| | - Cristina Daia
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.D.); (M.O.); (A.S.); (A.I.); (Ș.B.)
- Neuromuscular Rehabilitation Clinic Division, Teaching Emergency Hospital” Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.A.); (C.P.)
| | - Florentina Carmen Firan
- Physical and Rehabilitation Medicine & Balneology Clinic Division, Teaching Emergency Hospital of the Ilfov County, 022113 Bucharest, Romania;
| | - Mihaela Oprea
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.D.); (M.O.); (A.S.); (A.I.); (Ș.B.)
- Neuromuscular Rehabilitation Clinic Division, Teaching Emergency Hospital” Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.A.); (C.P.)
| | - Aura Spinu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.D.); (M.O.); (A.S.); (A.I.); (Ș.B.)
- Neuromuscular Rehabilitation Clinic Division, Teaching Emergency Hospital” Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.A.); (C.P.)
| | - Cristina Popescu
- Neuromuscular Rehabilitation Clinic Division, Teaching Emergency Hospital” Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.A.); (C.P.)
| | - Anca Ionescu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.D.); (M.O.); (A.S.); (A.I.); (Ș.B.)
| | - Ștefan Busnatu
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania; (C.D.); (M.O.); (A.S.); (A.I.); (Ș.B.)
| | - Constantin Munteanu
- Neuromuscular Rehabilitation Clinic Division, Teaching Emergency Hospital” Bagdasar-Arseni”, 041915 Bucharest, Romania; (A.A.); (C.P.)
- Faculty of Medical Bioengineering, University of Medicine and Pharmacy” Grigore T. Popa”, 700115 Iași, Romania
| |
Collapse
|
48
|
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
|
49
|
Wrzosek A, Gałecka S, Żochowska M, Olszewska A, Kulawiak B. Alternative Targets for Modulators of Mitochondrial Potassium Channels. Molecules 2022; 27:299. [PMID: 35011530 PMCID: PMC8746388 DOI: 10.3390/molecules27010299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial potassium channels control potassium influx into the mitochondrial matrix and thus regulate mitochondrial membrane potential, volume, respiration, and synthesis of reactive oxygen species (ROS). It has been found that pharmacological activation of mitochondrial potassium channels during ischemia/reperfusion (I/R) injury activates cytoprotective mechanisms resulting in increased cell survival. In cancer cells, the inhibition of these channels leads to increased cell death. Therefore, mitochondrial potassium channels are intriguing targets for the development of new pharmacological strategies. In most cases, however, the substances that modulate the mitochondrial potassium channels have a few alternative targets in the cell. This may result in unexpected or unwanted effects induced by these compounds. In our review, we briefly present the various classes of mitochondrial potassium (mitoK) channels and describe the chemical compounds that modulate their activity. We also describe examples of the multidirectional activity of the activators and inhibitors of mitochondrial potassium channels.
Collapse
Affiliation(s)
- Antoni Wrzosek
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.W.); (S.G.); (M.Ż.)
| | - Shur Gałecka
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.W.); (S.G.); (M.Ż.)
| | - Monika Żochowska
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.W.); (S.G.); (M.Ż.)
| | - Anna Olszewska
- Department of Histology, Medical University of Gdansk, 1a Debinki, 80-211 Gdansk, Poland;
| | - Bogusz Kulawiak
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.W.); (S.G.); (M.Ż.)
| |
Collapse
|
50
|
Luján R, Merchán-Pérez A, Soriano J, Martín-Belmonte A, Aguado C, Alfaro-Ruiz R, Moreno-Martínez AE, DeFelipe J. Neuron Class and Target Variability in the Three-Dimensional Localization of SK2 Channels in Hippocampal Neurons as Detected by Immunogold FIB-SEM. Front Neuroanat 2022; 15:781314. [PMID: 34975419 PMCID: PMC8715088 DOI: 10.3389/fnana.2021.781314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/19/2021] [Indexed: 11/22/2022] Open
Abstract
Small-conductance calcium-activated potassium (SK) channels are crucial for learning and memory. However, many aspects of their spatial organization in neurons are still unknown. In this study, we have taken a novel approach to answering these questions combining a pre-embedding immunogold labeling with an automated dual-beam electron microscope that integrates focused ion beam milling and scanning electron microscopy (FIB/SEM) to gather 3D map ultrastructural and biomolecular information simultaneously. Using this new approach, we evaluated the number and variability in the density of extrasynaptic SK2 channels in 3D reconstructions from six dendritic segments of excitatory neurons and six inhibitory neurons present in the stratum radiatum of the CA1 region of the mouse. SK2 immunoparticles were observed throughout the surface of hippocampal neurons, either scattered or clustered, as well as at intracellular sites. Quantitative volumetric evaluations revealed that the extrasynaptic SK2 channel density in spines was seven times higher than in dendritic shafts and thirty-five times higher than in interneurons. Spines showed a heterogeneous population of SK2 expression, some spines having a high SK2 content, others having a low content and others lacking SK2 channels. SK2 immunonegative spines were significantly smaller than those immunopositive. These results show that SK2 channel density differs between excitatory and inhibitory neurons and demonstrates a large variability in the density of SK2 channels in spines. Furthermore, we demonstrated that SK2 expression was associated with excitatory synapses, but not with inhibitory synapses in CA1 pyramidal cells. Consequently, regulation of excitability and synaptic plasticity by SK2 channels is expected to be neuron class- and target-specific. These data show that immunogold FIB/SEM represent a new powerful EM tool to correlate structure and function of ion channels with nanoscale resolution.
Collapse
Affiliation(s)
- Rafael Luján
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Angel Merchán-Pérez
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
| | - Joaquim Soriano
- CRIB-Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Alejandro Martín-Belmonte
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Carolina Aguado
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Rocío Alfaro-Ruiz
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Ana Esther Moreno-Martínez
- Synaptic Structure Laboratory, Instituto de Investigación en Discapacidades Neurológicas (IDINE), Departamento de Ciencias Médicas, Facultad de Medicina, Universidad Castilla-La Mancha, Albacete, Spain
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain.,Instituto Cajal (CSIC), Madrid, Spain
| |
Collapse
|